US20040171539A1 - Regulation of human protein kinase-like protein - Google Patents

Regulation of human protein kinase-like protein Download PDF

Info

Publication number
US20040171539A1
US20040171539A1 US10/471,762 US47176204A US2004171539A1 US 20040171539 A1 US20040171539 A1 US 20040171539A1 US 47176204 A US47176204 A US 47176204A US 2004171539 A1 US2004171539 A1 US 2004171539A1
Authority
US
United States
Prior art keywords
polypeptide
protein
seq
polynucleotide
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/471,762
Other versions
US7148050B2 (en
Inventor
Alex Smolyar
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bayer Pharma AG
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/471,762 priority Critical patent/US7148050B2/en
Assigned to BAYER AKTIENGESELLSCHAFT reassignment BAYER AKTIENGESELLSCHAFT ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SMOLYAR, ALEX
Publication of US20040171539A1 publication Critical patent/US20040171539A1/en
Assigned to BAYER HEALTHCARE AG reassignment BAYER HEALTHCARE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BAYER AKTIENGESELLSCHAFT
Application granted granted Critical
Publication of US7148050B2 publication Critical patent/US7148050B2/en
Assigned to BAYER SCHERING PHARMA AG reassignment BAYER SCHERING PHARMA AG MERGER (SEE DOCUMENT FOR DETAILS). Assignors: BAYER HEALTHCARE AG
Adjusted expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1205Phosphotransferases with an alcohol group as acceptor (2.7.1), e.g. protein kinases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the invention relates to the regulation of a human protein kinase-like protein.
  • Protein kinases play critical roles in the regulation of biochemical and morphological changes associated with cellular growth and division. D'Urso et al., Science 250, 786-91, 1990; Birchmeier et al., Bioessays 15, 185-89, 1993); U.S. Pat. No. 6,200,770. They serve as growth factor receptors and signal transducers and have been implicated in cellular transformation and malignancy. Hunter et al., Cell 70, 375-87, 1992; Posada et al., Mol. Biol. Cell 3, 583-92, 1992; Hunter et al., Cell 79, 573-82, 1994.
  • protein kinases have been shown to participate in the transmission of signals from growth-factor receptors (Sturgill et al., Nature 344, 715-18, 1988; Gomez et al., Nature 353, 170-73, 1991), control of entry of cells into mitosis (Nurse et al., Nature 344, 503-08, 1990; Maller, Curr. Opin. Cell Biol. 3, 269-75, 1991), and regulation of actin bundling (Husain-Chishti et al., Nature 334, 718-21, 1988).
  • Protein kinases can be divided into two main groups based on either amino acid sequence similarity or specificity for either serine/threonine or tyrosine residues. A small number of dual-specificity kinases are structurally like the serine/threonine-specific group. Within the broad classification, kinases can be further sub-divided into families whose members share a higher degree of catalytic domain amino acid sequence identity and also have similar biochemical properties. Most protein kinase family members also share structural features outside the kinase domain that reflect their particular cellular roles. These include regulatory domains that control kinase activity or interaction with other proteins. Hanks et al., Science 241, 42-52, 1988).
  • One embodiment of the invention is a proteine kinase-like protein polypeptide comprising an amino acid sequence selected from the group consisting of:
  • amino acid sequences which are at least about 35% identical to
  • amino acid sequences which are at least about 35% identical to
  • Yet another embodiment of the invention is a method of screening for agents which decrease extracellular matrix degradation.
  • a test compound is contacted with a proteine kinase-like protein polypeptide comprising an amino acid sequence selected from the group consisting of:
  • amino acid sequences which are at least about 35% identical to
  • amino acid sequences which are at least about 50% identical to
  • Binding between the test compound and the proteine kinase-like protein polypeptide is detected.
  • a test compound which binds to the proteine kinase-like protein polypeptide is thereby identified as a potential agent for decreasing extracellular matrix degradation.
  • the agent can work by decreasing the activity of the proteine kinase-like protein.
  • Another embodiment of the invention is a method of screening for agents which decrease extracellular matrix degradation.
  • a test compound is contacted with a polynucleotide encoding a proteine kinase-like protein polypeptide, wherein the polynucleotide comprises a nucleotide sequence selected from the group consisting of:
  • nucleotide sequences which are at least about 50% identical to
  • nucleotide sequences which are at least about 50% identical to
  • nucleotide sequences which are at least about 50% identical to
  • Binding of the test compound to the polynucleotide is detected.
  • a test compound which binds to the polynucleotide is identified as a potential agent for decreasing extracellular matrix degradation.
  • the agent can work by decreasing the amount of the proteine kinase-like protein through interacting with the proteine kinase-like protein mRNA.
  • Another embodiment of the invention is a method of screening for agents which regulate extracellular matrix degradation.
  • a test compound is contacted with a proteine kinase-like protein polypeptide comprising an amino acid sequence selected from the group consisting of:
  • amino acid sequences which are at least about 35% identical to
  • amino acid sequences which are at least about 35% identical to
  • a proteine kinase-like protein activity of the polypeptide is detected.
  • a test compound which increases proteine kinase-like protein activity of the polypeptide relative to proteine kinase-like protein activity in the absence of the test compound is thereby identified as a potential agent for increasing extracellular matrix degradation.
  • a test compound which decreases proteine kinase-like protein activity of the polypeptide relative to proteine kinase-like protein activity in the absence of the test compound is thereby identified as a potential agent for decreasing extracellular matrix degradation.
  • Yet another embodiment of the invention is a method of screening for agents which decrease extracellular matrix degradation.
  • a test compound is contacted with a proteine kinase-like protein product of a polynucleotide which comprises a nucleotide sequence selected from the group consisting of:
  • nucleotide sequences which are at least about 50% identical to
  • nucleotide sequences which are at least about 50% identical to
  • nucleotide sequences which are at least about 50% identical to
  • Binding of the test compound to the proteine kinase-like protein product is detected.
  • a test compound which binds to the proteine kinase-like protein product is thereby identified as a potential agent for decreasing extracellular matrix degradation.
  • Still another embodiment of the invention is a method of reducing extracellular matrix degradation.
  • a cell is contacted with a reagent which specifically binds to a polynucleotide encoding a proteine kinase-like protein polypeptide or the product encoded by the polynucleotide, wherein the polynucleotide comprises a nucleotide sequence selected from the group consisting of:
  • nucleotide sequences which are at least about 50% identical to
  • nucleotide sequences which are at least about 50% identical to
  • nucleotide sequences which are at least about 50% identical to
  • Proteine kinase-like protein activity in the cell is thereby decreased.
  • the invention thus provides a human protein kinase-like protein that can be used to identify test compounds that may act, for example, as activators or inhibitors at the enzyme's active site.
  • Human protein kinase-like protein and fragments thereof also are useful in raising specific antibodies that can block the enzyme and effectively reduce its activity.
  • FIG. 1 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:1).
  • FIG. 2 shows the amino acid sequence deduced from the DNA-sequence of FIG. 1 (SEQ ID NO:2).
  • FIG. 3 shows the amino acid sequence of the protein identified by swiss
  • FIG. 4 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:4).
  • FIG. 5 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:5).
  • FIG. 6 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:6).
  • FIG. 7 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:7).
  • FIG. 8 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:8).
  • FIG. 9 shows the amino acid sequence deduced from the DNA-sequence of FIG. 8 (SEQ ID NO:9).
  • FIG. 10 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:10).
  • FIG. 11 shows the BLASTP-alignment of 423 (SEQ ID NO:2) against swiss
  • FIG. 12 shows the HMMPFAM—alignment of 423 (SEQ ID NO:2) against pfam
  • FIG. 13 shows the relative expression of human protein kinase-like protein mRNA in cancer cells and tissue.
  • FIG. 14 shows the relative expression of protein kinase-like protein mRNA in tissue relevant for obesity and diabetes.
  • FIG. 15 shows the copy number of protein kinase-like protein mRNA in tissue relevant for obesity and diabetes.
  • FIG. 16 shows the relative expression profile of protein kinase-like protein mRNA in various human tissues (bodymap).
  • the invention relates to an isolated polynucleotide from the group consisting of:
  • amino acid sequences which are at least about 35% identical to the amino acid sequence shown in SEQ ID NO: 2;
  • amino acid sequences which are at least about 35% identical to the amino acid sequence shown in SEQ ID NO: 9 and;
  • a novel protein kinase-like protein can be used in therapeutic methods to treat cancer, a CNS disorder, COPD, obesity, diabetes, or a cardiovascular disorder.
  • Human protein kinase-like protein comprises the amino acid sequence shown in SEQ ID NOS:2 and 9.
  • a coding sequence for human protein kinase-like protein is shown in SEQ ID NOS: 1, 8 and 10. This sequence is located on chromosome 15.
  • Related ESTs (SEQ ID NOS:4-7) are expressed in lymphoma and germinal center B cells.
  • Human protein kinase-like protein is 31% identical over 287 amino acids to swiss
  • Pfam homology search results support the identification of this protein as a eukaryotic protein kinase. Its likely function as a tyrosine kinase class m protein also is supported by the results of a BLOCKS database search. Consensus patterns are underlined in FIG. 1; the active site Asp residue and the ATP-binding Lys residue are highlighted in bold.
  • Human protein kinase-like protein of the invention is expected to be useful for the same purposes as previously identified protein kinase-like protein enzymes. Human protein kinase-like protein is believed to be useful in therapeutic methods to treat disorders such as cancer, CNS disorders, COPD, obesity, diabetes, and cardiovascular disorders. Human protein kinase-like protein also can be used to screen for human protein kinase-like protein activators and inhibitors.
  • Human protein kinase-like polypeptides comprise at least 6, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, or 286 contiguous amino acids selected from the amino acid sequence shown in SEQ ID NO:2 or a biologically active variant thereof, as defined below.
  • Human protein kinase-like polypeptides comprise at least 6, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, 700, 725, 750, 775, 800, 825, 850, 875, 900, 925, 950, 975, 1000, 1025, 1050, 1075, 1100, 1125, 1150, 1175, 1200, 1225, 1250, 1275, 1300, 1325, 1350, 1375 or 1394 contiguous amino acids selected from the amino acid sequence shown in SEQ ID NO:9 or a biologically active variant thereof, as defined below.
  • a protein kinase-like polypeptide of the invention therefore can be a portion of a protein kinase-like protein, a full-length protein kinase-like protein, or a fusion protein comprising all or a portion of a protein kinase-like protein.
  • Human protein kinase-like polypeptide variants which are biologically active, e.g., retain enzymatic activity, also are human protein kinase-like polypeptides.
  • naturally or non-naturally occurring human protein kinase-like polypeptide variants have amino acid sequences which are at least about 32, 35, 40, 45, 50, 55, 60, 65, or 70, preferably about 75, 80, 85, 90, 96, 96, 98, or 99% identical to the amino acid sequence shown in SEQ ID NOS:2 and 9 or a fragment thereof.
  • Percent identity between a putative human protein kinase-like polypeptide variant and an amino acid sequence of SEQ ID NO:2 or 9 is determined by conventional methods. See, for example, Altschul et al., Bull. Math. Bio. 48:603 (1986), and Henikoff & Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1992). Briefly, two amino acid sequences are aligned to optimize the alignment scores using a gap opening penalty of 10, a gap extension penalty of 1, and the “BLOSUM62” scoring matrix of Henikoff & Henikoff, 1992.
  • the “FASTA” similarity search algorithm of Pearson & Lipman is a suitable protein alignment method for examining the level of identity shared by an amino acid sequence disclosed herein and the amino acid sequence of a putative variant.
  • the FASTA algorithm is described by Pearson & Lipman, Proc. Nat'l Acad. Sci. USA 85:2444(1988), and by Pearson, Meth. Enzymol. 183:63 (1990).
  • the trimmed initial regions are examined to determine whether the regions can be joined to form an approximate alignment with gaps.
  • the highest scoring regions of the two amino acid sequences are aligned using a modification of the Needleman-Wunsch-Sellers algorithm (Needleman & Wunsch, J. Mol. Biol. 48:444 (1970); Sellers, SIAM J. Appl. Math. 26:787 (1974)), which allows for amino acid insertions and deletions.
  • FASTA can also be used to determine the sequence identity of nucleic acid molecules using a ratio as disclosed above.
  • the ktup value can range between one to six, preferably from three to six, most preferably three, with other parameters set as default.
  • Variations in percent identity can be due, for example, to amino acid substitutions, insertions, or deletions.
  • Amino acid substitutions are defined as one for one amino acid replacements. They are conservative in nature when the substituted amino acid has similar structural and/or chemical properties. Examples of conservative replacements are substitution of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine.
  • Amino acid insertions or deletions are changes to or within an amino acid sequence. They typically fall in the range of about 1 to 5 amino acids. Guidance in determining which amino acid residues can be substituted, inserted, or deleted without abolishing biological or immunological activity of a human protein kinase-like polypeptide can be found using computer programs well known in the art, such as DNASTAR software.
  • the invention additionally, encompasses protein kinase-like polypeptides that are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications can be carried out by known techniques including, but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH 4 , acetylation, formylation, oxidation, reduction, metabolic synthesis in the presence of tunicamycin, etc.
  • Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of prokaryotic host cell expression.
  • the protein kinase-like polypeptides may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
  • the invention also provides chemically modified derivatives of protein kinase-like polypeptides that may provide additional advantages such as increased solubility, stability and circulating time of the polypeptide, or decreased immunogenicity (see U.S. Pat. No. 4,179,337).
  • the chemical moieties for derivitization can be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol, and the like.
  • the polypeptides can be modified at random or predetermined positions within the molecule and can include one, two, three, or more attached chemical moieties.
  • Fusion proteins are useful for generating antibodies against protein kinase-like polypeptide amino acid sequences and for use in various assay systems. For example, fusion proteins can be used to identify proteins that interact with portions of a protein kinase-like polypeptide. Protein affinity chromatography or library-based assays for protein-protein interactions, such as the yeast two-hybrid or phage display systems, can be used for this purpose. Such methods are well known in the art and also can be used as drug screens.
  • a protein kinase-like polypeptide fusion protein comprises two polypeptide segments fused together by means of a peptide bond.
  • the first polypeptide segment comprises at least 6, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, or 286 contiguous amino acids of SEQ D NOS:2 or of a biologically active variant, such as those described above.
  • the first polypeptide segment comprises at least 6, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, 700, 725, 750, 775, 800, 825, 850, 875, 900, 925, 950, 975, 1000, 1025, 1050, 1075, 1100, 1125, 1150, 1175, 1200, 1225, 1250, 1275, 1300, 1325, 1350, 1375 or 1394 contiguous amino acids of SEQ ID NOS:2 or of a biologically active variant, such as those described above.
  • the first polypeptide segment also can comprise full-length protein kinase-like protein.
  • the second polypeptide segment can be a full-length protein or a protein fragment.
  • Proteins commonly used in fusion protein construction include 13-galactosidase, P-glucuronidase, green fluorescent protein (GFP), autofluorescent proteins, including blue fluorescent protein (BFP), glutathione-S-transferase (GST), luciferase, horseradish peroxidase (HRP), and chloramphenicol acetyltransferase (CAT).
  • epitope tags are used in fusion protein constructions, including histidine (His) tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, VSV-G tags, and thioredoxin (Trx) tags.
  • Other fusion constructions can include maltose binding protein (MBP), S-tag, Lex a DNA binding domain (DBD) fusions, GALA DNA binding domain fusions, and herpes simplex virus (HSV) BP16 protein fusions.
  • a fusion protein also can be engineered to contain a cleavage site located between the protein kinase-like polypeptide-encoding sequence and the heterologous protein sequence, so that the protein kinase-like polypeptide can be cleaved and purified away from the heterologous moiety.
  • a fusion protein can be synthesized chemically, as is known in the art.
  • a fusion protein is produced by covalently linking two polypeptide segments or by standard procedures in the art of molecular biology.
  • Recombinant DNA methods can be used to prepare fusion proteins, for example, by making a DNA construct which comprises coding sequences selected from SEQ ID NOS: 1, 8 and 10 in proper reading frame with nucleotides encoding the second polypeptide segment and expressing the DNA construct in a host cell, as is known in the art.
  • kits for constructing fusion proteins are available from companies such as Promega Corporation (Madison, Wis.), Stratagene (La Jolla, Calif.), CLONTECH (Mountain View, Calif.), Santa Cruz Biotechnology (Santa Cruz, Calif.), MBL International Corporation (MIC; Watertown, Mass.), and Quantum Biotechnologies (Montreal, Canada; 1-888-DNA-KITS).
  • Species homologs of human protein kinase-like polypeptide can be obtained using protein kinase-like polypeptide polynucleotides (described below) to make suitable probes or primers for screening cDNA expression libraries from other species, such as mice, monkeys, or yeast, identifying cDNAs which encode homologs of protein kinase-like polypeptide, and expressing the cDNAs as is known in the art.
  • a protein kinase-like polynucleotide can be single- or double-stranded and comprises a coding sequence or the complement of a coding sequence for a protein kinase-like polypeptide.
  • a coding sequence for human protein kinase-like protein is shown in SEQ ID NOS: 1, 8 and 10.
  • nucleotide sequences encoding human protein kinase-like polypeptides as well as homologous nucleotide sequences which are at least about 50, 55, 60, 65, 70, preferably about 75, 90, 96, 98, or 99% identical to the nucleotide sequence shown in SEQ ID NOS: 1, 8 and 10 or its complement also are protein kinase-like polynucleotides. Percent sequence identity between the sequences of two polynucleotides is determined using computer programs such as ALIGN which employ the FASTA algorithm, using an affine gap search with a gap open penalty of ⁇ 12 and a gap extension penalty of ⁇ 2.
  • cDNA Complementary DNA
  • species homologs, and variants of protein kinase-like polynucleotides that encode biologically active protein kinase-like polypeptides also are protein kinase-like polynucleotides.
  • Polynucleotide fragments comprising at least 8, 9, 10, 11, 12, 15, 20, or 25 contiguous nucleotides of SEQ ID NOS: 1, 8 and 10 or its complement also are protein kinase-like polynucleotides. These fragments can be used, for example, as hybridization probes or as antisense oligonucleotides.
  • variants and homologs of the protein kinase-like polynucleotides described above also are protein kinase-like polynucleotides.
  • homologous protein kinase-like polynucleotide sequences can be identified by hybridization of candidate polynucleotides to known protein kinase-like polynucleotides under stringent conditions, as is known in the art. For example, using the following wash conditions—2 ⁇ SSC (0.3 M NaCl, 0.03 M sodium citrate, pH 7.0), 0.1% SDS, room temperature twice, 30 minutes each; then 2 ⁇ SSC, 0.1% SDS, 50° C.
  • homologous sequences can be identified which contain at most about 25-30% basepair mismatches. More preferably, homologous nucleic acid strands contain 15-25% basepair mismatches, even more preferably 5-15% basepair mismatches.
  • Species homologs of the protein kinase-like polynucleotides disclosed herein also can be identified by making suitable probes or primers and screening cDNA expression libraries from other species, such as mice, monkeys, or yeast.
  • Human variants of protein kinase-like polynucleotides can be identified, for example, by screening human cDNA expression libraries. It is well known that the T m of a double-stranded DNA decreases by 1-1.5° C. with every 1% decrease in homology (Bonner et al., J.
  • Variants of human protein kinase-like polynucleotides or protein kinase-like polynucleotides of other species can therefore be identified by hybridizing a putative homologous protein kinase-like polynucleotide with a polynucleotide having a nucleotide sequence of SEQ ID NOS: 1, 8 and 10 or the complement thereof to form a test hybrid.
  • the melting temperature of the test hybrid is compared with the melting temperature of a hybrid comprising polynucleotides having perfectly complementary nucleotide sequences, and the number or percent of basepair mismatches within the test hybrid is calculated.
  • Nucleotide sequences which hybridize to protein kinase-like polynucleotides or their complements following stringent hybridization and/or wash conditions also are protein kinase-like polynucleotides.
  • Stringent wash conditions are well known and understood in the art and are disclosed, for example, in Sambrook et al., M OLECULAR C LONING : A L ABORATORY M ANUAL, 2d ed., 1989, at pages 9.50-9.51.
  • T m a combination of temperature and salt concentration should be chosen that is approximately 12-20° C. below the calculated T m of the hybrid under study.
  • the T m of a hybrid between a protein kinase-like polynucleotide having a nucleotide sequence shown in SEQ ID NOS: 1, 8 and 10 or the complement thereof and a polynucleotide sequence which is at least about 50, preferably about 75, 90, 96, or 98% identical to one of those nucleotide sequences can be calculated, for example, using the equation of Bolton and McCarthy, Proc. Natl. Acad. Sci. U.S.A. 48, 1390 (1962):
  • T m 81.5 ° C.- 16.6(log 10 [Na + ])+0.41(% G+C )-0.63(% formamide )-600 /l ),
  • Stringent wash conditions include, for example, 4 ⁇ SSC at 65° C., or 50% formamide, 4 ⁇ SSC at 42° C., or 0.5 ⁇ SSC, 0.1% SDS at 65° C.
  • Highly stringent wash conditions include, for example, 0.2 ⁇ SSC at 65° C.
  • a protein kinase-like polynucleotide can be isolated free of other cellular components such as membrane components, proteins, and lipids.
  • Polynucleotides can be made by a cell and isolated using standard nucleic acid purification techniques, or synthesized using an amplification technique, such as the polymerase chain reaction (PCR), or by using an automatic synthesizer. Methods for isolating polynucleotides are routine and are known in the art. Any such technique for obtaining a polynucleotide can be used to obtain isolated protein kinase-like polynucleotides.
  • restriction enzymes and probes can be used to isolate polynucleotide fragments, which comprise protein kinase-like protein nucleotide sequences.
  • Isolated polynucleotides are in preparations that are free or at least 70, 80, or 90% free of other molecules.
  • Human protein kinase-like cDNA molecules can be made with standard molecular biology techniques, using protein kinase-like mRNA as a template. Human protein kinase-like cDNA molecules can thereafter be replicated using molecular biology techniques known in the art and disclosed in manuals such as Sambrook et al. (1989). An amplification technique, such as PCR, can be used to obtain additional copies of polynucleotides of the invention, using either human genomic DNA or cDNA as a template.
  • the partial sequence disclosed herein can be used to identify the corresponding full length gene from which it was derived.
  • the partial sequence can be nick-translated or end-labeled with 32 P using polynucleotide kinase using labeling methods known to those with skill in the art (B ASIC M ETHODS IN M OLECULAR B IOLOGY , Davis et al., eds., Elsevier Press, N.Y., 1986).
  • a lambda library prepared from human tissue can be directly screened with the labeled sequences of interest or the library can be converted en masse to pBluescript (Stratagene Cloning Systems, La Jolla, Calif. 92037) to facilitate bacterial colony screening (see Sambrook et al., M OLECULAR C LONING : A L ABORATORY M ANUAL , Cold Spring Harbor Laboratory Press (1989, pg. 1.20).
  • Positive cDNA clones are analyzed to determine the amount of additional sequence they contain using PCR with one primer from the partial sequence and the other primer from the vector.
  • Clones with a larger vector-insert PCR product than the original partial sequence are analyzed by restriction digestion and DNA sequencing to determine whether they contain an insert of the same size or similar as the mRNA size determined from Northern blot Analysis.
  • the complete sequence of the clones can be determined, for example after exonuclease III digestion (McCombie et al., Methods 3, 33-40, 1991). A series of deletion clones are generated, each of which is sequenced. The resulting overlapping sequences are assembled into a single contiguous sequence of high redundancy (usually three to five overlapping sequences at each nucleotide position), resulting in a highly accurate final sequence.
  • PCR-based methods can be used to extend the nucleic acid sequences disclosed herein to detect upstream sequences such as promoters and regulatory elements.
  • restriction-site PCR uses universal primers to retrieve unknown sequence adjacent to a known locus (Sarkar, PCR Methods Applic. 2, 318-322, 1993). Genomic DNA is first amplified in the presence of a primer to a linker sequence and a primer specific to the known region. The amplified sequences are then subjected to a second round of PCR with the same linker primer and another specific primer internal to the first one. Products of each round of PCR are transcribed with an appropriate RNA polymerase and sequenced using reverse transcriptase.
  • Inverse PCR also can be used to amplify or extend sequences using divergent primers based on a known region (Triglia et al., Nucleic Acids Res. 16, 8186, 1988). Primers can be designed using commercially available software, such as OLIGO 4.06 Primer Analysis software (National Biosciences Inc., Madison, Minn.), to be 22-30 nucleotides in length, to have a GC content of 50% or more, and to anneal to the target sequence at temperatures about 68-72° C. The method uses several restriction enzymes to generate a suitable fragment in the known region of a gene. The fragment is then circularized by intramolecular ligation and used as a PCR template.
  • capture PCR which involves PCR amplification of DNA fragments adjacent to a known sequence in human and yeast artificial chromosome DNA (Lagerstrom et al., PCR Methods Applic. 1, 111-119, 1991).
  • multiple restriction enzyme digestions and ligations also can be used to place an engineered double-stranded sequence into an unknown fragment of the DNA molecule before performing PCR.
  • Randomly-primed libraries are preferable, in that they will contain more sequences which contain the 5′ regions of genes. Use of a randomly primed library may be especially preferable for situations in which an oligo d(T) library does not yield a full-length cDNA. Genomic libraries can be useful for extension of sequence into 5′ non-transcribed regulatory regions.
  • capillary electrophoresis systems can be used to analyze the size or confirm the nucleotide sequence of PCR or sequencing products.
  • capillary sequencing can employ flowable polymers for electrophoretic separation, four different fluorescent dyes (one for each nucleotide) that are laser activated, and detection of the emitted wavelengths by a charge coupled device camera.
  • Output/light intensity can be converted to electrical signal using appropriate software (e.g. GENOTYPER and Sequence NAVIGATOR, Perkin Elmer), and the entire process from loading of samples to computer analysis and electronic data display can be computer controlled.
  • Capillary electrophoresis is especially preferable for the sequencing of small pieces of DNA that might be present in limited amounts in a particular sample.
  • Human protein kinase-like polypeptides can be obtained, for example, by purification from human or other cells, by expression of protein kinase-like polynucleotides, or by direct chemical synthesis.
  • Human protein kinase-like polypeptides can be purified from any cell that expresses the polypeptide, including host cells that have been transfected with protein kinase-like protein expression constructs.
  • a purified protein kinase-like polypeptide is separated from other compounds that normally associate with the protein kinase-like polypeptide in the cell, such as certain proteins, carbohydrates, or lipids, using methods well-known in the art. Such methods include, but are not limited to, size exclusion chromatography, ammonium sulfate fractionation, ion exchange chromatography, affinity chromatography, and preparative gel electrophoresis.
  • a preparation of purified protein kinase-like polypeptides is at least 80% pure; preferably, the preparations are 90%, 95%, or 99% pure. Purity of the preparations can be assessed by any means known in the art, such as SDS-polyacrylamide gel electrophoresis.
  • the polynucleotide can be inserted into an expression vector that contains the necessary elements for the transcription and translation of the inserted coding sequence.
  • Methods that are well known to those skilled in the art can be used to construct expression vectors containing sequences encoding protein kinase-like polypeptides and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described, for example, in Sambrook et al. (1989) and in Ausubel et al., C URRENT P ROTOCOLS IN M OLECULAR B IOLOGY , John Wiley & Sons, New York, N.Y., 1989.
  • a variety of expression vector/host systems can be utilized to contain and express sequences encoding a protein kinase-like polypeptide.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors, insect cell systems infected with virus expression vectors (e.g., baculovirus), plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids), or animal cell systems.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors
  • yeast transformed with yeast expression vectors insect cell systems infected with virus expression vectors (e.g., baculovirus), plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic
  • control elements or regulatory sequences are those non-translated regions of the vector—enhancers, promoters, 5′ and 3′ untranslated regions—which interact with host cellular proteins to carry out transcription and translation. Such elements can vary in their strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, can be used. For example, when cloning in bacterial systems, inducible promoters such as the hybrid lacZ promoter of the BLUESCRIPT phagemid (Stratagene, LaJolla, Calif.) or pSPORT1 plasmid (Life Technologies) and the like can be used. The baculovirus polyhedrin promoter can be used in insect cells.
  • Promoters or enhancers derived from the genomes of plant cells e.g., heat shock, RUBISCO, and storage genes
  • plant viruses e.g., viral promoters or leader sequences
  • promoters from mammalian genes or from mammalian viruses are preferable. If it is necessary to generate a cell line that contains multiple copies of a nucleotide sequence encoding a protein kinase-like polypeptide, vectors based on SV40 or EBV can be used with an appropriate selectable marker.
  • a number of expression vectors can be selected depending upon the use intended for the protein kinase-like polypeptide. For example, when a large quantity of a protein kinase-like polypeptide is needed for the induction of antibodies, vectors which direct high level expression of fusion proteins that are readily purified can be used. Such vectors include, but are not limited to, multifunctional E. coli cloning and expression vectors such as BLUESCRIPT (Stratagene).
  • a sequence encoding the protein kinase-like polypeptide can be ligated into the vector in frame with sequences for the amino-terminal Met and the subsequent 7 residues of ⁇ -galactosidase so that a hybrid protein is produced.
  • pIN vectors Van Heeke & Schuster, J. Biol. Chem. 264, 5503-5509, 1989
  • pGEX vectors Promega, Madison, Wis.
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione.
  • Proteins made in such systems can be designed to include heparin, thrombin, or factor Xa protease cleavage sites so that the cloned polypeptide of interest can be released from the GST moiety at will.
  • yeast Saccharomyces cerevisiae a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH can be used.
  • constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH.
  • sequences encoding protein kinase-like polypeptides can be driven by any of a number of promoters.
  • viral promoters such as the 35S and 19S promoters of CaMV can be used alone or in combination with the omega leader sequence from TMV (Takamatsu, EMBO J. 6, 307-311, 1987).
  • plant promoters such as the small subunit of RUBISCO or heat shock promoters can be used (Coruzzi et al., EMBO J. 3, 1671-1680, 1984; Broglie et al., Science 224, 838-843, 1984; Winter et al., Results Probl. Cell Differ.
  • constructs can be introduced into plant cells by direct DNA transformation or by pathogen-mediated transfection.
  • pathogen-mediated transfection Such techniques are described in a number of generally available reviews (e.g., Hobbs or Murray, in M C G RAW H ILL Y EARBOOK OF S CIENCE AND T ECHNOLOGY , McGraw Hill, New York, N.Y., pp. 191-196,1992).
  • An insect system also can be used to express a protein kinase-like polypeptide.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae.
  • Sequences encoding protein kinase-like polypeptides can be cloned into a non-essential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of protein kinase-like polypeptides will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein.
  • the recombinant viruses can then be used to infect S. frugiperda cells or Trichoplusia larvae in which protein kinase-like polypeptides can be expressed (Engelhard et al., Proc. Nat. Acad. Sci. 91, 3224-3227, 1994).
  • a number of viral-based expression systems can be used to express protein kinase-like polypeptides in mammalian host cells.
  • sequences encoding protein kinase-like polypeptides can be ligated into an adenovirus transcription/translation complex comprising the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome can be used to obtain a viable virus that is capable of expressing a protein kinase-like polypeptide in infected host cells (Logan & Shenk, Proc. Natl. Acad. Sci. 81, 3655-3659, 1984).
  • transcription enhancers such as the Rous sarcoma virus (RSV) enhancer, can be used to increase expression in mammalian host cells.
  • RSV Rous sarcoma virus
  • HACs Human artificial chromosomes
  • 6M to 10M are constructed and delivered to cells via conventional delivery methods (e.g., liposomes, polycationic amino polymers, or vesicles).
  • Specific initiation signals also can be used to achieve more efficient translation of sequences encoding protein kinase-like polypeptides. Such signals include the ATG initiation codon and adjacent sequences. In cases where sequences encoding a protein kinase-like polypeptide, its initiation codon, and upstream sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a fragment thereof, is inserted, exogenous translational control signals (including the ATG initiation codon) should be provided. The initiation codon should be in the correct reading frame to ensure translation of the entire insert. Exogenous translational elements and initiation codons can be of various origins, both natural and synthetic. The efficiency of expression can be enhanced by the inclusion of enhancers which are appropriate for the particular cell system which is used (see Scharf et al., Results Probl. Cell Differ. 20, 125-162, 1994).
  • a host cell strain can be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein kinase-like polypeptide in the desired fashion.
  • modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation.
  • Post-translational processing which cleaves a “prepro” form of the polypeptide also can be used to facilitate correct insertion, folding and/or function.
  • Different host cells that have specific cellular machinery and characteristic mechanisms for post-translational activities e.g., CHO, HeLa, MDCK, HEK293, and WI38
  • ATCC American Type Culture Collection
  • Stable expression is preferred for long-term, high-yield production of recombinant proteins.
  • cell lines which stably express protein kinase-like polypeptides can be transformed using expression vectors which can contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells can be allowed to grow for 1-2 days in an enriched medium before they are switched to a selective medium.
  • the purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells which successfully express the introduced protein kinase-like protein sequences.
  • Resistant clones of stably transformed cells can be proliferated using tissue culture techniques appropriate to the cell type. See, for example, A NIMAL C ELL C ULTURE , R. I. Freshney, ed., 1986.
  • herpes simplex virus thymidine kinase (Wigler et al., Cell 11, 223-32, 1977) and adenine phosphoribosyltransferase (Lowy et al., Cell 22, 817-23, 1980) genes which can be employed in tk ⁇ or aprt ⁇ (cells, respectively.
  • antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection.
  • dhfr confers resistance to methotrexate (Wigler et al., Proc. Natl. Acad. Sci.
  • npt confers resistance to the aminoglycosides, neomycin and G-418 (Colbere-Garapin et al., J. Mol. Biol. 150, 1-14, 1981), and als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (Murray, 1992, supra). Additional selectable genes have been described. For example, trpB allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine (Hartman & Mulligan, Proc. Natl. Acad. Sci. 85, 8047-51, 1988).
  • Visible markers such as anthocyanins, ⁇ -glucuronidase and its substrate GUS, and luciferase and its substrate luciferin, can be used to identify transformants and to quantify the amount of transient or stable protein expression attributable to a specific vector system (Rhodes et al., Methods Mol. Biol. 55, 121-131, 1995).
  • marker gene expression suggests that the protein kinase-like polynucleotide is also present, its presence and expression may need to be confirmed. For example, if a sequence encoding a protein kinase-like polypeptide is inserted within a marker gene sequence, transformed cells containing sequences that encode a protein kinase-like polypeptide can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding a protein kinase-like polypeptide under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the protein kinase-like polynucleotide.
  • host cells which contain a protein kinase-like polynucleotide and which express a protein kinase-like polypeptide can be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations and protein bioassay or immunoassay techniques that include membrane, solution, or chip-based technologies for the detection and/or quantification of nucleic acid or protein.
  • the presence of a polynucleotide sequence encoding a protein kinase-like polypeptide can be detected by DNA-DNA or DNA-RNA hybridization or amplification using probes or fragments or fragments of polynucleotides encoding a protein kinase-like polypeptide.
  • Nucleic acid amplification-based assays involve the use of oligonucleotides selected from sequences encoding a protein kinase-like polypeptide to detect transformants that contain a protein kinase-like polynucleotide.
  • a variety of protocols for detecting and measuring the expression of a protein kinase-like polypeptide, using either polyclonal or monoclonal antibodies specific for the polypeptide, are known in the art. Examples include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence activated cell sorting (FACS).
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • FACS fluorescence activated cell sorting
  • a two-site, monoclonal-based immunoassay using monoclonal antibodies reactive to two non-interfering epitopes on a protein kinase-like polypeptide can be used, or a competitive binding assay can be employed.
  • a wide variety of labels and conjugation techniques are known by those skilled in the art and can be used in various nucleic acid and amino acid assays.
  • Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding protein kinase-like polypeptides include oligolabeling, nick translation, end-labeling, or PCR amplification using a labeled nucleotide.
  • sequences encoding a protein kinase-like polypeptide can be cloned into a vector for the production of an mRNA probe.
  • RNA probes are known in the art, are commercially available, and can be used to synthesize RNA probes in vitro by addition of labeled nucleotides and an appropriate RNA polymerase such as T7, T3, or SP6. These procedures can be conducted using a variety of commercially available kits (Amersham Pharmacia Biotech, Promega, and US Biochemical). Suitable reporter molecules or labels which can be used for ease of detection include radionuclides, enzymes, and fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
  • Host cells transformed with nucleotide sequences encoding a protein kinase-like polypeptide can be cultured under conditions suitable for the expression and recovery of the protein from cell culture.
  • the polypeptide produced by a transformed cell can be secreted or contained intracellularly depending on the sequence and/or the vector used.
  • expression vectors containing polynucleotides which encode protein kinase-like polypeptides can be designed to contain signal sequences which direct secretion of soluble protein kinase-like polypeptides through a prokaryotic or eukaryotic cell membrane or which direct the membrane insertion of membrane-bound protein kinase-like polypeptide.
  • purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, Wash.).
  • cleavable linker sequences such as those specific for Factor Xa or enterokinase (Invitrogen, San Diego, Calif.) between the purification domain and the protein kinase-like polypeptide also can be used to facilitate purification.
  • One such expression vector provides for expression of a fusion protein containing a protein kinase-like polypeptide and 6 histidine residues preceding a thioredoxin or an enterokinase cleavage site. The histidine residues facilitate purification by IMAC (immobilized metal ion affinity chromatography, as described in Porath et al., Prot. Exp. Purif.
  • enterokinase cleavage site provides a means for purifying the protein kinase-like polypeptide from the fusion protein.
  • Vectors that contain fusion proteins are disclosed in Kroll et al., DNA Cell Biol. 12,441453,1993.
  • Sequences encoding a protein kinase-like polypeptide can be synthesized, in whole or in part, using chemical methods well known in the art (see Caruthers et al., Nucl. Acids Res. Symp. Ser. 215-223, 1980; Horn et al. Nucl. Acids Res. Symp. Ser. 225-232, 1980).
  • a protein kinase-like polypeptide itself can be produced using chemical methods to synthesize its amino acid sequence, such as by direct peptide synthesis using solid-phase techniques (Merrifield, J. Am. Chem. Soc. 85, 2149-2154, 1963; Roberge et al., Science 269, 202-204, 1995).
  • Protein synthesis can be performed using manual techniques or by automation. Automated synthesis can be achieved, for example, using Applied Biosystems 431A Peptide Synthesizer (Perkin Elmer). Optionally, fragments of protein kinase-like polypeptides can be separately synthesized and combined using chemical methods to produce a full-length molecule.
  • the newly synthesized peptide can be substantially purified by preparative high performance liquid chromatography (e.g., Creighton, P ROTEINS : S TRUCTURES AND M OLECULAR P RINCIPLES , WH Freeman and Co., New York, N.Y., 1983).
  • the composition of a synthetic protein kinase-like polypeptide can be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure; see Creighton, supra). Additionally, any portion of the amino acid sequence of the protein kinase-like polypeptide can be altered during direct synthesis and/or combined using chemical methods with sequences from other proteins to produce a variant polypeptide or a fusion protein.
  • codons preferred by a particular prokaryotic or eukaryotic host can be selected to increase the rate of protein expression or to produce an RNA transcript having desirable properties, such as a half-life that is longer than that of a transcript generated from the naturally occurring sequence.
  • nucleotide sequences disclosed herein can be engineered using methods generally known in the art to alter protein kinase-like polypeptide-encoding sequences for a variety of reasons, including but not limited to, alterations which modify the cloning, processing, and/or expression of the polypeptide or mRNA product.
  • DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides can be used to engineer the nucleotide sequences.
  • site-directed mutagenesis can be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, introduce mutations, and so forth.
  • Antibody as used herein includes intact immunoglobulin molecules, as well as fragments thereof, such as Fab, F(ab′) 2 , and Fv, which are capable of binding an epitope of a protein kinase-like polypeptide.
  • epitopes typically, at least 6, 8, 10, or 12 contiguous amino acids are required to form an epitope.
  • epitopes which involve non-contiguous amino acids may require more, e.g., at least 15, 25, or 50 amino acids.
  • An antibody which specifically binds to an epitope of a protein kinase-like polypeptide can be used therapeutically, as well as in immunochemical assays, such as Western blots, ELISAs, radioimmunoassays, immunohistochemical assays, immunoprecipitations, or other immunochemical assays known in the art.
  • immunochemical assays such as Western blots, ELISAs, radioimmunoassays, immunohistochemical assays, immunoprecipitations, or other immunochemical assays known in the art.
  • Various immunoassays can be used to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays are well known in the art. Such immunoassays typically involve the measurement of complex formation between an immunogen and an antibody that specifically binds to the immunogen.
  • an antibody which specifically binds to a protein kinase-like polypeptide provides a detection signal at least 5-, 10-, or 20-fold higher than a detection signal provided with other proteins when used in an immunochemical assay.
  • antibodies which specifically bind to protein kinase-like polypeptides do not detect other proteins in immunochemical assays and can immunoprecipitate a protein kinase-like polypeptide from solution.
  • Human protein kinase-like polypeptides can be used to immunize a mammal, such as a mouse, rat, rabbit, guinea pig, monkey, or human, to produce polyclonal antibodies.
  • a protein kinase-like polypeptide can be conjugated to a carrier protein, such as bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin.
  • a carrier protein such as bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin.
  • various adjuvants can be used to increase the immunological response.
  • adjuvants include, but are not limited to, Freund's adjuvant, mineral gels (e.g., aluminum hydroxide), and surface active substances (e.g.
  • BCG Bacilli Calmette-Guerin
  • Corynebacterium parvum are especially useful.
  • Monoclonal antibodies that specifically bind to a protein kinase-like polypeptide can be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These techniques include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique (Kohler et al., Nature 256, 495-497, 1985; Kozbor et al., J. Immunol. Methods 81, 31-42, 1985; Cote et al., Proc. Natl. Acad. Sci. 80, 2026-2030, 1983; Cole et al., Mol. Cell Biol. 62, 109-120, 1984).
  • chimeric antibodies the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used (Morrison et al., Proc Natl. Acad. Sci. 81, 6851-6855, 1984; Neuberger et al., Nature 312, 604-608, 1984; Takeda et al., Nature 314, 452-454, 1985).
  • Monoclonal and other antibodies also can be “humanized” to prevent a patient from mounting an immune response against the antibody when it is used therapeutically. Such antibodies may be sufficiently similar in sequence to human antibodies to be used directly in therapy or may require alteration of a few key residues.
  • rodent antibodies and human sequences can be minimized by replacing residues which differ from those in the human sequences by site directed mutagenesis of individual residues or by grating of entire complementarity determining regions.
  • humanized antibodies can be produced using recombinant methods, as described in GB2188638B.
  • Antibodies that specifically bind to a protein kinase-like polypeptide can contain antigen binding sites which are either partially or fully humanized, as disclosed in U.S. Pat. No. 5,565,332.
  • single chain antibodies can be adapted using methods known in the art to produce single chain antibodies that specifically bind to protein kinase-like polypeptides.
  • Antibodies with related specificity, but of distinct idiotypic composition can be generated by chain shuffling from random combinatorial immunoglobin libraries (Burton, Proc. Natl. Acad. Sci. 88, 11120-23, 1991).
  • Single-chain antibodies also can be constructed using a DNA amplification method, such as PCR, using hybridoma cDNA as a template (Thirion et al., 1996 , Eur. J. Cancer Prev. 5, 507-11).
  • Single-chain antibodies can be mono- or bispecific, and can be bivalent or tetravalent. Construction of tetravalent, bispecific single-chain antibodies is taught, for example, in Coloma & Morrison, 1997 , Nat. Biotechnol. 15, 159-63. Construction of bivalent, bispecific single-chain antibodies is taught in Mallender & Voss, 1994 , J. Biol. Chem. 269, 199-206.
  • a nucleotide sequence encoding a single-chain antibody can be constructed using manual or automated nucleotide synthesis, cloned into an expression construct using standard recombinant DNA methods, and introduced into a cell to express the coding sequence, as described below.
  • single-chain antibodies can be produced directly using, for example, filamentous phage technology (Verhaar et al., 1995 , Int. J. Cancer 61, 497-501; Nicholls et al., 1993 , J. Immunol. Meth. 165, 81-91).
  • Antibodies which specifically bind to protein kinase-like polypeptides also can be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature (Orlandi et al., Proc. Natl. Acad. Sci. 86, 3833-3837, 1989; Winter et al., Nature 349, 293-299, 1991).
  • chimeric antibodies can be constructed as disclosed in WO 93/03151.
  • Binding proteins which are derived from immunoglobulins and which are multivalent and multispecific, such as the “diabodies” described in WO 94/13804, also can be prepared.
  • Antibodies according to the invention can be purified by methods well known in the art. For example, antibodies can be affinity purified by passage over a column to which a protein kinase-like polypeptide is bound. The bound antibodies can then be eluted from the column using a buffer with a high salt concentration.
  • Antisense oligonucleotides are nucleotide sequences that are complementary to a specific DNA or RNA sequence. Once introduced into a cell, the complementary nucleotides combine with natural sequences produced by the cell to form complexes and block either transcription or translation.
  • an antisense oligonucleotide is at least 11 nucleotides in length, but can be at least 12, 15, 20, 25, 30, 35, 40, 45, or 50 or more nucleotides long. Longer sequences also can be used.
  • Antisense oligonucleotide molecules can be provided in a DNA construct and introduced into a cell as described above to decrease the level of protein kinase-like gene products in the cell.
  • Antisense oligonucleotides can be deoxyribonucleotides, ribonucleotides, or a combination of both. Oligonucleotides can be synthesized manually or by an automated synthesizer, by covalently linking the 5′ end of one nucleotide with the 3′ end of another nucleotide with non-phosphodiester internucleotide linkages such alkyl-phosphonates, phosphorothioates, phosphorodithioates, alkylphosphonothioates, alkylphosphonates, phosphoramidates, phosphate esters, carbamates, acetamidate, carboxymethyl esters, carbonates, and phosphate triesters. See Brown, Meth. Mol. Biol. 20, 1-8, 1994; Sonveaux, Meth. Mol. Biol. 26, 1-72, 1994; Uhlmann et al., Chem. Rev. 90, 543-583,1990.
  • Modifications of protein kinase-like gene expression can be obtained by designing antisense oligonucleotides that will form duplexes to the control, 5′, or regulatory regions of the protein kinase-like gene. Oligonucleotides derived from the transcription initiation site, e.g., between positions ⁇ 10 and +10 from the start site, are preferred. Similarly, inhibition can be achieved using “triple helix” base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or chaperons.
  • An antisense oligonucleotide also can be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.
  • Antisense oligonucleotides which comprise, for example, 2, 3, 4, or 5 or more stretches of contiguous nucleotides which are precisely complementary to a protein kinase-like polynucleotide, each separated by a stretch of contiguous nucleotides which are not complementary to adjacent protein kinase-like protein nucleotides, can provide sufficient targeting specificity for protein kinase-like mRNA.
  • each stretch of complementary contiguous nucleotides is at least 4, 5, 6, 7, or 8 or more nucleotides in length.
  • Non-complementary intervening sequences are preferably 1, 2, 3, or 4 nucleotides in length.
  • One skilled in the art can easily use the calculated melting point of an antisense-sense pair to determine the degree of mismatching which will be tolerated between a particular antisense oligonucleotide and a particular protein kinase-like polynucleotide sequence.
  • Antisense oligonucleotides can be modified without affecting their ability to hybridize to a protein kinase-like polynucleotide. These modifications can be internal or at one or both ends of the antisense molecule.
  • internucleoside phosphate linkages can be modified by adding cholesteryl or diamine moieties with varying numbers of carbon residues between the amino groups and terminal ribose.
  • Modified bases and/or sugars such as arabinose instead of ribose, or a 3′,5′-substituted oligonucleotide in which the 3′ hydroxyl group or the 5′ phosphate group are substituted, also can be employed in a modified antisense oligonucleotide.
  • modified oligonucleotides can be prepared by methods well known in the art. See, e.g., Agrawal et al., Trends Biotechnol. 10, 152-158, 1992; Uhlmann et al., Chem. Rev. 90, 543-584, 1990; Uhlmann et al., Tetrahedron. Lett. 215, 3539-3542, 1987.
  • Ribozymes are RNA molecules with catalytic activity. See, e.g., Cech, Science 236, 1532-1539; 1987; Cech, Ann. Rev. Biochem. 59, 543-568; 1990, Cech, Curr. Opin. Struct. Biol. 2, 605-609; 1992, Couture & Stinchcomb, Trends Genet. 12, 510-515, 1996. Ribozymes can be used to inhibit gene function by cleaving an RNA sequence, as is known in the art (e.g., Haseloff et al., U.S. Pat. No. 5,641,673).
  • ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • Examples include engineered hammerhead motif ribozyme molecules that can specifically and efficiently catalyze endonucleolytic cleavage of specific nucleotide sequences.
  • the coding sequence of a protein kinase-like polynucleotide can be used to generate ribozymes that will specifically bind to mRNA transcribed from the protein kinase-like polynucleotide.
  • Methods of designing and constructing ribozymes which can cleave other RNA molecules in trans in a highly sequence specific manner have been developed and described in the art (see Haseloff et al. Nature 334, 585-591, 1988).
  • the cleavage activity of ribozymes can be targeted to specific RNAs by engineering a discrete “hybridization” region into the ribozyme.
  • the hybridization region contains a sequence complementary to the target RNA and thus specifically hybridizes with the target (see, for example, Gerlach et al., EP 321,201).
  • Specific ribozyme cleavage sites within a protein kinase-like protein RNA target can be identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences: GUA, GUU, and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides corresponding to the region of the target RNA containing the cleavage site can be evaluated for secondary structural features which may render the target inoperable. Suitability of candidate protein kinase-like protein RNA targets also can be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays.
  • hybridizing and cleavage regions of the ribozyme can be integrally related such that upon hybridizing to the target RNA through the complementary regions, the catalytic region of the ribozyme can cleave the target.
  • Ribozymes can be introduced into cells as part of a DNA construct. Mechanical methods, such as microinjection, liposome-mediated transfection, electroporation, or calcium phosphate precipitation, can be used to introduce a ribozyme-containing DNA construct into cells in which it is desired to decrease protein kinase-like protein expression. Alternatively, if it is desired that the cells stably retain the DNA construct, the construct can be supplied on a plasmid and maintained as a separate element or integrated into the genome of the cells, as is known in the art.
  • a ribozyme-encoding DNA construct can include transcriptional regulatory elements, such as a promoter element, an enhancer or UAS element, and a transcriptional terminator signal, for controlling transcription of ribozymes in the cells.
  • ribozymes can be engineered so that ribozyme expression will occur in response to factors that induce expression of a target gene. Ribozymes also can be engineered to provide an additional level of regulation, so that destruction of mRNA occurs only when both a ribozyme and a target gene are induced in the cells.
  • genes whose products interact with human protein kinase-like protein may represent genes that are differentially expressed in disorders including, but not limited to, cancer, CNS disorders, COPD, obesity, diabetes, and cardiovascular disorders
  • genes may represent genes that are differentially regulated in response to manipulations relevant to the progression or treatment of such diseases. Additionally, such genes may have a temporally modulated expression, increased or decreased at different stages of tissue or organism development. A differentially expressed gene may also have its expression modulated under control versus experimental conditions. In addition, the human protein kinase-like gene or gene product may itself be tested for differential expression.
  • the degree to which expression differs in a normal versus a diseased state need only be large enough to be visualized via standard characterization techniques such as differential display techniques.
  • standard characterization techniques such as differential display techniques.
  • Other such standard characterization techniques by which expression differences may be visualized include but are not limited to, quantitative RT (reverse transcriptase), PCR, and Northern analysis.
  • RNA samples are obtained from tissues of experimental subjects and from corresponding tissues of control subjects. Any RNA isolation technique that does not select against the isolation of mRNA may be utilized for the purification of such RNA samples. See, for example, Ausubel et al., ed., C URRENT P ROTOCOLS IN M OLECULAR B IOLOGY , John Wiley & Sons, Inc. New York, 1987-1993. Large numbers of tissue samples may readily be processed using techniques well known to those of skill in the art, such as, for example, the single-step RNA isolation process of Chomczynski, U.S. Pat. No. 4,843,155.
  • Transcripts within the collected RNA samples that represent RNA produced by differentially expressed genes are identified by methods well known to those of skill in the art. They include, for example, differential screening (Tedder et al., Proc. Natl. Acad. Sci. U.S.A. 85, 208-12, 1988), subtractive hybridization (Hedrick et al., Nature 308, 149-53; Lee et al., Proc. Natl. Acad. Sci. U.S.A. 88, 2825, 1984), and, preferably, differential display (Liang & Pardee, Science 257, 967-71, 1992; U.S. Pat. No. 5,262,311).
  • the differential expression information may itself suggest relevant methods for the treatment of disorders involving the human protein kinase-like protein.
  • treatment may include a modulation of expression of the differentially expressed genes and/or the gene encoding the human protein kinase-like protein.
  • the differential expression information may indicate whether the expression or activity of the differentially expressed gene or gene product or the human protein kinase-like gene or gene product are up-regulated or down-regulated.
  • the invention provides assays for screening test compounds that bind to or modulate the activity of a protein kinase-like polypeptide or a protein kinase-like polynucleotide.
  • a test compound preferably binds to a protein kinase-like polypeptide or polynucleotide. More preferably, a test compound decreases or increases enzyme activity by at least about 10, preferably about 50, more preferably about 75, 90, or 100% relative to the absence of the test compound.
  • Test compounds can be pharmacologic agents already known in the art or can be compounds previously unknown to have any pharmacological activity.
  • the compounds can be naturally occurring or designed in the laboratory. They can be isolated from microorganisms, animals, or plants, and can be produced recombinantly, or synthesized by chemical methods known in the art. If desired, test compounds can be obtained using any of the numerous combinatorial library methods known in the art, including but not limited to, biological libraries, spatially addressable parallel solid phase or solution phase libraries, synthetic library methods requiring deconvolution, the “one-bead one-compound” library method, and synthetic library methods using affinity chromatography selection.
  • the biological library approach is limited to polypeptide libraries, while the other four approaches are applicable to polypeptide, non-peptide oligomer, or small molecule libraries of compounds. See Lam, Anticancer Drug Des. 12, 145, 1997.
  • Test compounds can be screened for the ability to bind to protein kinase-like polypeptides or polynucleotides or to affect protein kinase-like protein activity or protein kinase-like gene expression using high throughput screening.
  • high throughput screening many discrete compounds can be tested in parallel so that large numbers of test compounds can be quickly screened.
  • the most widely established techniques utilize 96-well microfiter plates. The wells of the microtiter plates typically require assay volumes that range from 50 to 500 ⁇ l.
  • many instruments, materials, pipettors, robotics, plate washers, and plate readers are commercially available to fit the 96-well format.
  • free format assays or assays that have no physical barrier between samples, can be used.
  • an assay using pigment cells (melanocytes) in a simple homogeneous assay for combinatorial peptide libraries is described by Jayawickreme et al., Proc. Natl. Acad. Sci. U.S.A. 19, 1614-18 (1994).
  • the cells are placed under agarose in petri dishes, then beads that carry combinatorial compounds are placed on the surface of the agarose.
  • the combinatorial compounds are partially released the compounds from the beads. Active compounds can be visualized as dark pigment areas because, as the compounds diffuse locally into the gel matrix, the active compounds cause the cells to change colors.
  • test samples are placed in a porous matrix.
  • One or more assay components are then placed within, on top of, or at the bottom of a matrix such as a gel, a plastic sheet, a filter, or other form of easily manipulated solid support.
  • a matrix such as a gel, a plastic sheet, a filter, or other form of easily manipulated solid support.
  • the test compound is preferably a small molecule that binds to and occupies, for example, the active site of the protein kinase-like polypeptide, such that normal biological activity is prevented.
  • small molecules include, but are not limited to, small peptides or peptide-like molecules.
  • either the test compound or the protein kinase-like polypeptide can comprise a detectable label, such as a fluorescent, radioisotopic, chemiluminescent, or enzymatic label, such as horseradish peroxidase, alkaline phosphatase, or luciferase.
  • a detectable label such as a fluorescent, radioisotopic, chemiluminescent, or enzymatic label, such as horseradish peroxidase, alkaline phosphatase, or luciferase.
  • Detection of a test compound that is bound to the protein kinase-like polypeptide can then be accomplished, for example, by direct counting of radio-emmission, by scintillation counting, or by determining conversion of an appropriate substrate to a detectable product.
  • binding of a test compound to a protein kinase-like polypeptide can be determined without labeling either of the interactants.
  • a microphysiometer can be used to detect binding of a test compound with a protein kinase-like polypeptide.
  • a microphysiometer e.g., CytosensorTM
  • a microphysiometer is an analytical instrument that measures the rate at which a cell acidifies its environment using a light-addressable potentiometric sensor (LAPS). Changes in this acidification rate can be used as an indicator of the interaction between a test compound and a protein kinase-like polypeptide (McConnell et al., Science 257, 1906-1912, 1992).
  • BIA Bimolecular Interaction Analysis
  • a protein kinase-like polypeptide can be used as a “bait protein” in a two-hybrid assay or three-hybrid assay (see, e.g., U.S. Pat. No. 5,283,317; Zervos et al., Cell 72, 223-232, 1993; Madura et al., J. Biol. Chem.
  • the two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains.
  • the assay utilizes two different DNA constructs.
  • polynucleotide encoding a protein kinase-like polypeptide can be fused to a polynucleotide encoding the DNA binding domain of a known transcription factor (e.g., GAL-4).
  • a DNA sequence that encodes an unidentified protein (“prey” or “sample” can be fused to a polynucleotide that codes for the activation domain of the known transcription factor.
  • the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ), which is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected, and cell colonies containing the functional transcription factor can be isolated and used to obtain the DNA sequence encoding the protein that interacts with the protein kinase-like polypeptide.
  • a reporter gene e.g., LacZ
  • either the protein kinase-like polypeptide (or polynucleotide) or the test compound can be bound to a solid support.
  • Suitable solid supports include, but are not limited to, glass or plastic slides, tissue culture plates, microtiter wells, tubes, silicon chips, or particles such as beads (including, but not limited to, latex, polystyrene, or glass beads).
  • Any method known in the art can be used to attach the enzyme polypeptide (or polynucleotide) or test compound to a solid support, including use of covalent and non-covalent linkages, passive absorption, or pairs of binding moieties attached respectively to the polypeptide (or polynucleotide) or test compound and the solid support.
  • Test compounds are preferably bound to the solid support in an array, so that the location of individual test compounds can be tracked. Binding of a test compound to a protein kinase-like polypeptide (or polynucleotide) can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and microcentrifuge tubes.
  • the protein kinase-like polypeptide is a fusion protein comprising a domain that allows the protein kinase-like polypeptide to be bound to a solid support.
  • glutathione-S-transferase fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, Mo.) or glutathione derivatized microtiter plates, which are then combined with the test compound or the test compound and the non-adsorbed protein kinase-like polypeptide; the mixture is then incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH).
  • Binding of the interactants can be determined either directly or indirectly, as described above. Alternatively, the complexes can be dissociated from the solid support before binding is determined.
  • Biotinylated protein kinase-like polypeptides (or polynucleotides) or test compounds can be prepared from biotin-NHS(N-hydroxysuccinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, Ill.) and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • biotinylation kit Pierce Chemicals, Rockford, Ill.
  • antibodies which specifically bind to a protein kinase-like polypeptide, polynucleotide, or a test compound, but which do not interfere with a desired binding site, such as the active site of the protein kinase-like polypeptide can be derivatized to the wells of the plate. Unbound target or protein can be trapped in the wells by antibody conjugation.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies which specifically bind to the protein kinase-like polypeptide or test compound, enzyme-linked assays which rely on detecting an activity of the protein kinase-like polypeptide, and SDS gel electrophoresis under non-reducing conditions.
  • Screening for test compounds which bind to a protein kinase-like polypeptide or polynucleotide also can be carried out in an intact cell. Any cell which comprises a protein kinase-like polypeptide or polynucleotide can be used in a cell-based assay system. A protein kinase-like polynucleotide can be naturally occurring in the cell or can be introduced using techniques such as those described above. Binding of the test compound to a protein kinase-like polypeptide or polynucleotide is determined as described above.
  • Test compounds can be tested for the ability to increase or decrease the protein kinase activity of a human protein kinase-like polypeptide. Protein kinase activity can be measured, for example, as described in U.S. Patent U.S. Pat. No. 6,194,186
  • Enzyme assays can be carried out after contacting either a purified protein kinase-like polypeptide, a cell membrane preparation, or an intact cell with a test compound.
  • a test compound that decreases a protein kinase activity of a protein kinase-like polypeptide by at least about 10, preferably about 50, more preferably about 75, 90, or 100% is identified as a potential therapeutic agent for decreasing protein kinase-like protein activity.
  • a test compound which increases a protein kinase activity of a human protein kinase-like polypeptide by at least about 10, preferably about 50, more preferably about 75, 90, or 100% is identified as a potential therapeutic agent for increasing human protein kinase-like protein activity.
  • test compounds that increase or decrease protein kinase-like gene expression are identified.
  • a protein kinase-like polynucleotide is contacted with a test compound, and the expression of an RNA or polypeptide product of the protein kinase-like polynucleotide is determined.
  • the level of expression of appropriate mRNA or polypeptide in the presence of the test compound is compared to the level of expression of mRNA or polypeptide in the absence of the test compound.
  • the test compound can then be identified as a modulator of expression based on this comparison.
  • test compound when expression of mRNA or polypeptide is greater in the presence of the test compound than in its absence, the test compound is identified as a stimulator or enhancer of the mRNA or polypeptide expression.
  • test compound when expression of the mRNA or polypeptide is less in the presence of the test compound than in its absence, the test compound is identified as an inhibitor of the mRNA or polypeptide expression.
  • the level of protein kinase-like mRNA or polypeptide expression in the cells can be determined by methods well known in the art for detecting mRNA or polypeptide. Either qualitative or quantitative methods can be used.
  • the presence of polypeptide products of a protein kinase-like polynucleotide can be determined, for example, using a variety of techniques known in the art, including immunochemical methods such as radioimmunoassay, Western blotting, and immunohistochemistry.
  • polypeptide synthesis can be determined in vivo, in a cell culture, or in an in vitro translation system by detecting incorporation of labeled amino acids into a protein kinase-like polypeptide.
  • Such screening can be carried out either in a cell-free assay system or in an intact cell.
  • Any cell that expresses a protein kinase-like polynucleotide can be used in a cell-based assay system.
  • the protein kinase-like polynucleotide can be naturally occurring in the cell or can be introduced using techniques such as those described above.
  • Either a primary culture or an established cell line, such as CHO or human embryonic kidney 293 cells, can be used.
  • compositions of the invention can comprise, for example, a protein kinase-like polypeptide, protein kinase-like polynucleotide, ribozymes or antisense oligonucleotides, antibodies which specifically bind to a protein kinase-like polypeptide, or mimetics, activators, or inhibitors of a protein kinase-like polypeptide activity.
  • compositions can be administered alone or in combination with at least one other agent, such as stabilizing compound, which can be administered in any sterile, biocompatible pharmaceutical carrier, including, but not limited to, saline, buffered saline, dextrose, and water.
  • agent such as stabilizing compound
  • the compositions can be administered to a patient alone, or in combination with other agents, drugs or hormones.
  • compositions of the invention can be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intraperitoneal, intranasal, parenteral, topical, sublingual, or rectal means.
  • Pharmaceutical compositions for oral administration can be formulated using pharmaceutically acceptable carriers well known in the art in dosages suitable for oral administration. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for ingestion by the patient.
  • compositions for oral use can be obtained through combination of active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are carbohydrate or protein fillers, such as sugars, including lactose, sucrose, mannitol, or sorbitol; starch from corn, wheat, rice, potato, or other plants; cellulose, such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethylcellulose; gums including arabic and tragacanth; and proteins such as gelatin and collagen.
  • disintegrating or solubilizing agents can be added, such as the cross-linked polyvinyl pyrrolidone, agar, alginic acid, or a salt thereof, such as sodium alginate.
  • Dragee cores can be used in conjunction with suitable coatings, such as concentrated sugar solutions, which also can contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • suitable coatings such as concentrated sugar solutions, which also can contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments can be added to the tablets or dragee coatings for product identification or to characterize the quantity of active compound, i.e., dosage.
  • compositions that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating, such as glycerol or sorbitol.
  • Push-fit capsules can contain active ingredients mixed with a filler or binders, such as lactose or starches, lubricants, such as talc or magnesium stearate, and, optionally, stabilizers.
  • the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid, or liquid polyethylene glycol with or without stabilizers.
  • compositions suitable for parenteral administration can be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiologically buffered saline.
  • Aqueous injection suspensions can contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • suspensions of the active compounds can be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Non-lipid polycationic amino polymers also can be used for delivery.
  • the suspension also can contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • compositions of the present invention can be manufactured in a manner that is known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes.
  • the pharmaceutical composition can be provided as a salt and can be formed with many acids, including but not limited to, hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms.
  • the preferred preparation can be a lyophilized powder which can contain any or all of the following: 1-50 mM histidine, 0.1%-2% sucrose, and 2-7% mannitol, at a pH range of 4.5 to 5.5, that is combined with buffer prior to use.
  • Human protein kinase-like protein can be regulated to treat cancer, CNS disorders, COPD, obesity, diabetes, and cardiovascular disorders.
  • Cancer is a disease fundamentally caused by oncogenic cellular transformation. There are several hallmarks of transformed cells that distinguish them from their normal counterparts and underlie the pathophysiology of cancer. These include uncontrolled cellular proliferation, unresponsiveness to normal death-inducing signals (immortalization), increased cellular motility and invasiveness, increased ability to recruit blood supply through induction of new blood vessel formation (angiogenesis), genetic instability, and dysregulated gene expression. Various combinations of these aberrant physiologies, along with the acquisition of drug-resistance frequently lead to an intractable disease state in which organ failure and patient death ultimately ensue.
  • Genes or gene fragments identified through genomics can readily be expressed in one or more heterologous expression systems to produce functional recombinant proteins. These proteins are characterized in vitro for their biochemical properties and then used as tools in high-throughput molecular screening programs to identify chemical modulators of their biochemical activities. Activators and/or inhibitors of target protein activity can be identified in this manner and subsequently tested in cellular and in vivo disease models for anti-cancer activity. Optimization of lead compounds with iterative testing in biological models and detailed pharmacokinetic and toxicological analyses form the basis for drug development and subsequent testing in humans.
  • Protein phosphorylation is an essential component in intracellular signaling, with diverse and crucial functions including mediation of cell proliferation, survival, apoptosis, differentiation, migration and attachment. It is regulated by the balance between the opposing activities of protein kinases and protein phosphatases. Protein phosphorylation is mainly mediated by two types of protein kinases—protein tyrosine kinases and protein serine/threonine kinases. A number of protein tyrosine kinases are encoded by proto-oncogenes or viral oncogenes, and are thus strongly implicated in cancer.
  • Protein serine/threonine kinases are known to play a role in intracellular signal transduction mediated by growth factors, cytokines, etc. inducing either cell proliferation, apoptosis or differentiation. Inhibitors of protein kinases are expected to provide efficacious therapeutic agents for the treatment of cancer.
  • Central and peripheral nervous system disorders also can be treated, such as primary and secondary disorders after brain injury, disorders of mood, anxiety disorders, disorders of thought and volition, disorders of sleep and wakefulness, diseases of the motor unit, such as neurogenic and myopathic disorders, neurodegenerative disorders such as Alzheimer's and Parkinson's disease, and processes of peripheral and chronic pain.
  • Pain that is associated with CNS disorders also can be treated by regulating the activity of human protein kinase-like protein. Pain which can be treated includes that associated with central nervous system disorders, such as multiple sclerosis, spinal cord injury, sciatica, failed back surgery syndrome, traumatic brain injury, epilepsy, Parkinson's disease, post-stroke, and vascular lesions in the brain and spinal cord (e.g., infarct, hemorrhage, vascular malformation).
  • central nervous system disorders such as multiple sclerosis, spinal cord injury, sciatica, failed back surgery syndrome, traumatic brain injury, epilepsy, Parkinson's disease, post-stroke, and vascular lesions in the brain and spinal cord (e.g., infarct, hemorrhage, vascular malformation).
  • Non-central neuropathic pain includes that associated with post mastectomy pain, reflex sympathetic dystrophy (RSD), trigeminal neuralgiaradioculopathy, post-surgical pain, HIV/AIDS related pain, cancer pain, metabolic neuropathies (e.g., diabetic neuropathy, vasculitic neuropathy secondary to connective tissue disease), paraneoplastic polyneuropathy associated, for example, with carcinoma of lung, or leukemia, or lymphoma, or carcinoma of prostate, colon or stomach, trigeminal neuralgia, cranial neuralgias, and post-herpetic neuralgia. Pain associated with cancer and cancer treatment also can be treated, as can headache pain (for example, migraine with aura, migraine without aura, and other migraine disorders), episodic and chronic tension-type headache, tension-type like headache, cluster headache, and chronic paroxysmal hemicrania.
  • headache pain for example, migraine with aura, migraine without aura, and other migraine disorders
  • episodic and chronic tension-type headache tension-type like headache, cluster headache, and chronic par
  • COPD chronic obstructive pulmonary (or airways) disease
  • COPD chronic obstructive pulmonary (or airways) disease
  • COPD chronic obstructive pulmonary (or airways) disease
  • COPD chronic obstructive pulmonary (or airways) disease
  • Emphysema is characterized by destruction of alveolar walls leading to abnormal enlargement of the air spaces of the lung.
  • Chronic bronchitis is defined clinically as the presence of chronic productive cough for three months in each of two successive years.
  • airflow obstruction is usually progressive and is only partially reversible. By far the most important risk factor for development of COPD is cigarette smoking, although the disease does occur in non-smokers.
  • Chronic inflammation of the airways is a key pathological feature of COPD (Senior & Shapiro, 1998).
  • the inflammatory cell population comprises increased numbers of macrophages, neutrophils, and CD8+ lymphocytes.
  • Inhaled irritants such as cigarette smoke, activate macrophages which are resident in the respiratory tract, as well as epithelial cells leading to release of chemokines (e.g., interleukin-8) and other chemotactic factors.
  • chemokines e.g., interleukin-8
  • These chemotactic factors act to increase the neutrophil/-monocyte trafficking from the blood into the lung tissue and airways.
  • Neutrophils and monocytes recruited into the airways can release a variety of potentially damaging mediators such as proteolytic enzymes and reactive oxygen species.
  • Matrix degradation and emphysema along with airway wall thickening, surfactant dysfunction, and mucus hypersecretion, all are potential sequelae of this
  • Obesity and overweight are defined as an excess of body fat relative to lean body mass. An increase in caloric intake or a decrease in energy expenditure or both can bring about this imbalance leading to surplus energy being stored as fat. Obesity is associated with important medical morbidities and an increase in mortality. The causes of obesity are poorly understood and may be due to genetic factors, environmental factors or a combination of the two to cause a positive energy balance. In contrast, anorexia and cachexia are characterized by an imbalance in energy intake versus energy expenditure leading to a negative energy balance and weight loss. Agents that either increase energy expenditure and/or decrease energy intake, absorption or storage would be useful for treating obesity, overweight, and associated comorbidities. Agents that either increase energy intake and/or decrease energy expenditure or increase the amount of lean tissue would be useful for treating cachexia, anorexia and wasting disorders.
  • This gene, translated proteins and agents which modulate this gene or portions of the gene or its products are useful for treating obesity, overweight, anorexia, cachexia, wasting disorders, appetite suppression, appetite enhancement, increases or decreases in satiety, modulation of body weight, and/or other eating disorders such as bulimia.
  • this gene translated proteins and agents which modulate this gene or portions of the gene or its products are useful for treating obesity/overweight-associated comorbidities including hypertension, type 2 diabetes, coronary artery disease, hyper-lipidemia, stroke, gallbladder disease, gout, osteoarthritis, sleep apnea and respiratory problems, some types of cancer including endometrial, breast, prostate, and colon cancer, thrombolic disease, polycystic ovarian syndrome, reduced fertility, complications of pregnancy, menstrual irregularities, hirsutism, stress incontinence, and depression.
  • obesity/overweight-associated comorbidities including hypertension, type 2 diabetes, coronary artery disease, hyper-lipidemia, stroke, gallbladder disease, gout, osteoarthritis, sleep apnea and respiratory problems, some types of cancer including endometrial, breast, prostate, and colon cancer, thrombolic disease, polycystic ovarian syndrome, reduced fertility, complications of pregnancy, menstrual irregularities, hir
  • Diabetes mellitus is a common metabolic disorder characterized by an abnormal elevation in blood glucose, alterations in lipids and abnormalities (complications) in the cardiovascular system, eye, kidney and nervous system. Diabetes is divided into two separate diseases: type 1 diabetes (juvenile onset), which results from a loss of cells which make and secrete insulin, and type 2 diabetes (adult onset), which is caused by a defect in insulin secretion and a defect in insulin action.
  • type 1 diabetes juvenile onset
  • type 2 diabetes adult onset
  • Type 1 diabetes is initiated by an autoimmune reaction that attacks the insulin secreting cells (beta cells) in the pancreatic islets.
  • Agents that prevent this reaction from occurring or that stop the reaction before destruction of the beta cells has been accomplished are potential therapies for this disease.
  • Other agents that induce beta cell proliferation and regeneration also are potential therapies.
  • Type II diabetes is the most common of the two diabetic conditions (6% of the population).
  • the defect in insulin secretion is an important cause of the diabetic condition and results from an inability of the beta cell to properly detect and respond to rises in blood glucose levels with insulin release.
  • Therapies that increase the response by the beta cell to glucose would offer an important new treatment for this disease.
  • the defect in insulin action in Type II diabetic subjects is another target for therapeutic intervention.
  • Agents that increase the activity of the insulin receptor in muscle, liver, and fat will cause a decrease in blood glucose and a normalization of plasma lipids.
  • the receptor activity can be increased by agents that directly stimulate the receptor or that increase the intracellular signals from the receptor.
  • Other therapies can directly activate the cellular end process, i.e. glucose transport or various enzyme systems, to generate an insulin-like effect and therefore a produce beneficial outcome. Because overweight subjects have a greater susceptibility to Type II diabetes, any agent that reduces body weight is a possible therapy.
  • Type I and Type diabetes can be treated with agents that mimic insulin action or that treat diabetic complications by reducing blood glucose levels.
  • agents that reduces new blood vessel growth can be used to treat the eye complications that develop in both diseases.
  • Cardiovascular diseases include the following disorders of the heart and the vascular system: congestive heart failure, myocardial infarction, ischemic diseases of the heart, all kinds of atrial and ventricular arrhythmias, hypertensive vascular diseases, and peripheral vascular diseases.
  • Heart failure is defined as a pathophysiologic state in which an abnormality of cardiac function is responsible for the failure of the heart to pump blood at a rate commensurate with the requirement of the metabolizing tissue. It includes all forms of pumping failure, such as high-output and low-output, acute and chronic, right-sided or left-sided, systolic or diastolic, independent of the underlying cause.
  • MI Myocardial infarction
  • Ischemic diseases are conditions in which the coronary flow is restricted resulting in a perfusion which is inadequate to meet the myocardial requirement for oxygen.
  • This group of diseases includes stable angina, unstable angina, and asymptomatic ischemia.
  • Arrhythmias include all forms of atrial and ventricular tachyarrhythmias (atrial tachycardia, atrial flutter, atrial fibrillation, atrio-ventricular reentrant tachycardia, preexcitation syndrome, ventricular tachycardia, ventricular flutter, and ventricular fibrillation), as well as bradycardic forms of arrhythmias.
  • vascular diseases include primary as well as all kinds of secondary arterial hypertension (renal, endocrine, neurogenic, others).
  • the disclosed gene and its product may be used as drug targets for the treatment of hypertension as well as for the prevention of all complications.
  • Peripheral vascular diseases are defined as vascular diseases in which arterial and/or venous flow is reduced resulting in an imbalance between blood supply and tissue oxygen demand. It includes chronic peripheral arterial occlusive disease (PAOD), acute arterial thrombosis and embolism, inflammatory vascular disorders, Raynaud's phenomenon, and venous disorders.
  • PAOD peripheral arterial occlusive disease
  • This invention further pertains to the use of novel agents identified by the screening assays described above. Accordingly, it is within the scope of this invention to use a test compound identified as described herein in an appropriate animal model.
  • an agent identified as described herein e.g., a modulating agent, an antisense nucleic acid molecule, a specific antibody, ribozyme, or a protein kinase-like polypeptide binding molecule
  • an agent identified as described herein can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent.
  • an agent identified as described herein can be used in an animal model to determine the mechanism of action of such an agent.
  • this invention pertains to uses of novel agents identified by the above-described screening assays for treatments as described herein.
  • a reagent which affects protein kinase-like protein activity can be administered to a human cell, either in vitro or in vivo, to reduce protein kinase-like protein activity.
  • the reagent preferably binds to an expression product of a human protein kinase-like gene. If the expression product is a protein, the reagent is preferably an antibody.
  • an antibody can be added to a preparation of stem cells that have been removed from the body. The cells can then be replaced in the same or another human body, with or without clonal propagation, as is known in the art.
  • the reagent is delivered using a liposome.
  • the liposome is stable in the animal into which it has been administered for at least about 30 minutes, more preferably for at least about 1 hour, and even more preferably for at least about 24 hours.
  • a liposome comprises a lipid composition that is capable of targeting a reagent, particularly a polynucleotide, to a particular site in an animal, such as a human.
  • the lipid composition of the liposome is capable of targeting to a specific organ of an animal, such as the lung, liver, spleen, heart brain, lymph nodes, and skin.
  • a liposome useful in the present invention comprises a lipid composition that is capable of fusing with the plasma membrane of the targeted cell to deliver its contents to the cell.
  • the transfection efficiency of a liposome is about 0.5 ⁇ g of DNA per 16 nmole of liposome delivered to about 10 6 cells, more preferably about 1.0 ⁇ g of DNA per 16 nmole of liposome delivered to about 10 6 cells, and even more preferably about 2.0 ⁇ g of DNA per 16 nmol of liposome delivered to about 10 6 cells.
  • a liposome is between about 100 and 500 nm, more preferably between about 150 and 450 nm, and even more preferably between about 200 and 400 nm in diameter.
  • Suitable liposomes for use in the present invention include those liposomes standardly used in, for example, gene delivery methods known to those of skill in the art. More preferred liposomes include liposomes having a polycationic lipid composition and/or liposomes having a cholesterol backbone conjugated to polyethylene glycol.
  • a liposome comprises a compound capable of targeting the liposome to a particular cell type, such as a cell-specific ligand exposed on the outer surface of the liposome.
  • a liposome with a reagent such as an antisense oligonucleotide or ribozyme can be achieved using methods that are standard in the art (see, for example, U.S. Pat. No. 5,705,151).
  • a reagent such as an antisense oligonucleotide or ribozyme
  • antibodies can be delivered to specific tissues in vivo using receptor-mediated targeted delivery.
  • Receptor-mediated DNA delivery techniques are taught in, for example, Findeis et al. Trends in Biotechnol. 11, 202-05 (1993); Chiou et al., G ENE T HERAPEUTICS : M ETHODS AND A PPLICATIONS OF D IRECT G ENE T RANSFER (J. A. Wolff, ed.) (1994); Wu & Wu, J. Biol. Chem. 263, 621-24 (1988); Wu et al., J. Biol. Chem. 269, 542-46 (1994); Zenke et al., Proc. Natl. Acad. Sci. U.S.A. 87, 3655-59 (1990); Wu et al., J. Biol. Chem. 266, 338-42 (1991).
  • a therapeutically effective dose refers to that amount of active ingredient which increases or decreases protein kinase-like protein activity relative to the protein kinase-like protein activity which occurs in the absence of the therapeutically effective dose.
  • the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rabbits, dogs, or pigs.
  • the animal model also can be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • Therapeutic efficacy and toxicity e.g., ED 50 (the dose therapeutically effective in 50% of the population) and LD 50 (the dose lethal to 50% of the population), can be determined by standard pharmaceutical procedures in cell cultures or experimental animals.
  • the dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD 50 /ED 50 .
  • compositions that exhibit large therapeutic indices are preferred.
  • the data obtained from cell culture assays and animal studies is used in formulating a range of dosage for human use.
  • the dosage contained in such compositions is preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage varies within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
  • the exact dosage will be determined by the practitioner, in light of factors related to the subject that requires treatment. Dosage and administration are adjusted to provide sufficient levels of the active ingredient or to maintain the desired effect. Factors that can be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions can be administered every 3 to 4 days, every week, or once every two weeks depending on the half-life and clearance rate of the particular formulation.
  • Normal dosage amounts can vary from 0.1 to 100,000 micrograms, up to a total dose of about 1 g, depending upon the route of administration.
  • Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art. Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc.
  • the reagent is a single-chain antibody
  • polynucleotides encoding the antibody can be constructed and introduced into a cell either ex vivo or in vivo using well-established techniques including, but not limited to, transferrin-polycation-mediated DNA transfer, transfection with naked or encapsulated nucleic acids, liposome-mediated cellular fusion, intracellular transportation of DNA-coated latex beads, protoplast fusion, viral infection, electroporation, “gene gun,” and DEAE- or calcium phosphate-mediated transfection.
  • Effective in vivo dosages of an antibody are in the range of about 5 ⁇ g to about 50 ⁇ g/kg, about 50 ⁇ g to about 5 mg/kg, about 100 ⁇ g to about 500 ⁇ g/kg of patient body weight, and about 200 to about 250 ⁇ g/kg of patient body weight.
  • effective in vivo dosages are in the range of about 100 ng to about 200 ng, 500 ng to about 50 mg, about 1 ⁇ g to about 2 mg, about 5 ⁇ g to about 500 ⁇ g, and about 20 ⁇ g to about 100 ⁇ g of DNA.
  • the reagent is preferably an antisense oligo-nucleotide or a ribozyme.
  • Polynucleotides that express antisense oligonucleotides or ribozymes can be introduced into cells by a variety of methods, as described above.
  • a reagent reduces expression of a protein kinase-like gene or the activity of a protein kinase-like polypeptide by at least about 10, preferably about 50, more preferably about 75, 90, or 100% relative to the absence of the reagent.
  • the effectiveness of the mechanism chosen to decrease the level of expression of a protein kinase-like gene or the activity of a protein kinase-like polypeptide can be assessed using methods well known in the art, such as hybridization of nucleotide probes to protein kinase-like protein-specific mRNA, quantitative RT-PCR, immunologic detection of a protein kinase-like polypeptide, or measurement of protein kinase-like protein activity.
  • any of the pharmaceutical compositions of the invention can be administered in combination with other appropriate therapeutic agents.
  • Selection of the appropriate agents for use in combination therapy can be made by one of ordinary skill in the art, according to conventional pharmaceutical principles.
  • the combination of therapeutic agents can act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
  • any of the therapeutic methods described above can be applied to any subject in need of such therapy, including, for example, mammals such as dogs, cats, cows, horses, rabbits, monkeys, and most preferably, humans.
  • Human protein kinase-like protein also can be used in diagnostic assays for detecting diseases and abnormalities or susceptibility to diseases and abnormalities related to the presence of mutations in the nucleic acid sequences that encode the enzyme. For example, differences can be determined between the cDNA or genomic sequence encoding protein kinase-like protein in individuals afflicted with a disease and in normal individuals. If a mutation is observed in some or all of the afflicted individuals but not in normal individuals, then the mutation is likely to be the causative agent of the disease.
  • Sequence differences between a reference gene and a gene having mutations can be revealed by the direct DNA sequencing method.
  • cloned DNA segments can be employed as probes to detect specific DNA segments.
  • the sensitivity of this method is greatly enhanced when combined with PCR.
  • a sequencing primer can be used with a double-stranded PCR product or a single-stranded template molecule generated by a modified PCR.
  • the sequence determination is performed by conventional procedures using radiolabeled nucleotides or by automatic sequencing procedures using fluorescent tags.
  • DNA sequence differences can be carried out by detection of alteration in electrophoretic mobility of DNA fragments in gels with or without denaturing agents. Small sequence deletions and insertions can be visualized, for example, by high resolution gel electrophoresis. DNA fragments of different sequences can be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g., Myers et al., Science 230, 1242, 1985).
  • Sequence changes at specific locations can also be revealed by nuclease protection assays, such as RNase and S 1 protection or the chemical cleavage method (e.g., Cotton et al., Proc. Natl. Acad. Sci. USA 85, 4397-4401, 1985).
  • nuclease protection assays such as RNase and S 1 protection or the chemical cleavage method (e.g., Cotton et al., Proc. Natl. Acad. Sci. USA 85, 4397-4401, 1985).
  • the detection of a specific DNA sequence can be performed by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes and Southern blotting of genomic DNA.
  • mutations can also be detected by in situ analysis.
  • Altered levels of protein kinase-like protein also can be detected in various tissues.
  • Assays used to detect levels of the receptor polypeptides in a body sample, such as blood or a tissue biopsy, derived from a host are well known to those of skill in the art and include radioimmunoassays, competitive binding assays, Western blot analysis, and ELISA assays.
  • the polynucleotide of SEQ ID NO: 1 is inserted into the expression vector pCEV4 and the expression vector pCEV4-proteine kinase-like protein polypeptide obtained is transfected into human embryonic kidney 293 cells. From these cells extracts are obtained and proteine kinase-like protein activity is assayed by measuring the incorporation of 32Pi into calf thymus H1 histone from [gamma-32P]ATP.
  • Cell extracts are incubated in a reaction mixture containing 20 mM Tris HCl at pH 7.5, 10 mM MgCl2, 20 ⁇ M ATP, 15-50 kBq of [gamma-32P]ATP, and 200 ⁇ g/ml H1 istone.
  • the incubation is carried out for 5 min at 30° C., and the phosphorylated proteins are seperated by SDS/PAGE and visualized and quanitated by measuring the intensity of photostimulated luminescene (PSL) using a Bio-Imaging Analyzer. It is shown that the polypeptide of SEQ ID NO: 2 has a proteine kinase-like protein activity.
  • the Pichia pastoris expression vector pPICZB (Invitrogen, San Diego, Calif.) is used to produce large quantities of recombinant human protein kinase-like polypeptides in yeast.
  • the protein kinase-like protein-encoding DNA sequence is derived from SEQ ID NOS: 1, 8 and 10.
  • the DNA sequence is modified by well known methods in such a way that it contains at its 5′-end an initiation codon and at its 3′-end an enterokinase cleavage site, a His6 reporter tag and a termination codon.
  • the yeast is cultivated under usual conditions in 5 liter shake flasks and the recombinantly produced protein isolated from the culture by affinity chromatography (Ni-NTA-Resin) in the presence of 8 M urea.
  • the bound polypeptide is eluted with buffer, pH 3.5, and neutralized. Separation of the polypeptide from the His6 reporter tag is accomplished by site-specific proteolysis using enterokinase (Invitrogen, San Diego, Calif.) according to manufacturer's instructions. Purified human protein kinase-like polypeptide is obtained.
  • Purified protein kinase-like polypeptides comprising a glutathione-S-transferase protein and absorbed onto glutathione-derivatized wells of 96-well microtiter plates are contacted with test compounds from a small molecule library at pH 7.0 in a physiological buffer solution.
  • Human protein kinase-like polypeptides comprise the amino acid sequence shown in SEQ ID NOS:2 and 9.
  • the test compounds comprise a fluorescent tag.
  • the samples are incubated for 5 minutes to one hour. Control samples are incubated in the absence of a test compound.
  • the buffer solution containing the test compounds is washed from the wells. Binding of a test compound to a protein kinase-like polypeptide is detected by fluorescence measurements of the contents of the wells. A test compound that increases the fluorescence in a well by at least 15% relative to fluorescence of a well in which a test compound is not incubated is identified as a compound which binds to a protein kinase-like polypeptide.
  • test compound is administered to a culture of human cells transfected with a protein kinase-like protein expression construct and incubated at 37° C. for 10 to 45 minutes.
  • a culture of the same type of cells that have not been transfected is incubated for the same time without the test compound to provide a negative control.
  • RNA is isolated from the two cultures as described in Chirgwin et al., Biochem. 18, 5294-99, 1979).
  • Northern blots are prepared using 20 to 30 ⁇ g total RNA and hybridized with a 32 P-labeled protein kinase-like protein-specific probe at 65° C. in Express-hyb (CLONTECH).
  • the probe comprises at least 11 contiguous nucleotides selected from the complement of SEQ ID NOS: 1, 8 and 10.
  • a test compound that decreases the protein kinase-like protein-specific signal relative to the signal obtained in the absence of the test compound is identified as an inhibitor of protein kinase-like gene expression.
  • test compound is administered to a culture of human cells transfected with a protein kinase-like protein expression construct and incubated at 37° C. for 10 to 45 minutes.
  • a culture of the same type of cells that have not been transfected is incubated for the same time without the test compound to provide a negative control.
  • protein kinase-like protein activity is measured using the method of U.S. Pat. No. 6,194,186.
  • a test compound which decreases the protein kinase-like protein activity of the protein kinase-like protein relative to the protein kinase-like protein activity in the absence of the test compound is identified as an inhibitor of protein kinase-like protein activity.
  • RT-PCR Reverse Transcription-Polymerase Chain Reaction
  • protein kinase-like protein is involved in cancer, expression is determined in the following tissues: adrenal gland, bone marrow, brain, cerebellum, colon, fetal brain, fetal liver, heart, kidney, liver, lung, mammary gland, pancreas, placenta, prostate, salivary gland, skeletal muscle, small intestine, spinal cord, spleen, stomach, testis, thymus, thyroid, trachea, uterus, and peripheral blood lymphocytes.
  • Expression in the following cancer cell lines also is determined: DU-145 (prostate), NCI-H125 (lung), HT-29 (colon), COLO-205 (colon), A-549 (lung), NCI—H460 (lung), HT-116 (colon), DLD-1 (colon), MDA-MD-231 (breast), LS 174T (colon), ZF-75 (breast), MDA-MN-435 (breast), HT-1080, MCF-7 (breast), and U87. Matched pairs of malignant and normal tissue from the same patient also are tested.
  • fetal and adult brain muscle, heart, lung, kidney, liver, thymus, testis, colon, placenta, trachea, pancreas, kidney, gastric mucosa, colon, liver, cerebellum, skin, cortex (Alzheimer's and normal), hypothalamus, cortex, amygdala, cerebellum, hippocampus, choroid, plexus, thalamus, and spinal cord.
  • the initial expression panel consists of RNA samples from respiratory tissues and inflammatory cells relevant to COPD: lung (adult and fetal), trachea, freshly isolated alveolar type II cells, cultured human bronchial epithelial cells, cultured small airway epithelial cells, cultured bronchial sooth muscle cells, cultured H441 cells (Clara-like), freshly isolated neutrophils and monocytes, and cultured monocytes (macrophage-like).
  • Body map profiling also is carried out, using total RNA panels purchased from Clontech.
  • the tissues are adrenal gland, bone marrow, brain, colon, heart, kidney, liver, lung, mammary gland, pancreas, prostate, salivary gland, skeletal muscle, small intestine, spleen, stomach, testis, thymus, trachea, thyroid, and uterus.
  • protein kinase-like protein is involved in the disease process of obesity, expression is determined in the following tissues: subcutaneous adipose tissue, mesenteric adipose tissue, adrenal gland, bone marrow, brain (cerebellum, spinal cord, cerebral cortex, caudate, medulla, substantia nigra, and putamen), colon, fetal brain, heart, kidney, liver, lung, mammary gland, pancreas, placenta, prostate, salivary gland, skeletal muscle small intestine, spleen, stomach, testes, thymus, thyroid trachea, and uterus.
  • tissues subcutaneous adipose tissue, mesenteric adipose tissue, adrenal gland, bone marrow, brain (cerebellum, spinal cord, cerebral cortex, caudate, medulla, substantia nigra, and putamen), colon, fetal brain, heart, kidney, liver, lung, mammary gland, pancre
  • Quantitative expression profiling is performed by the form of quantitative PCR analysis called “kinetic analysis” firstly described in Higuchi et al., BioTechnology 10, 413-17, 1992, and Higuchi et al., BioTechnology 11, 1026-30, 1993. The principle is that at any given cycle within the exponential phase of PCR, the amount of product is proportional to the initial number of template copies.
  • the amplification is performed in the presence of an internally quenched fluorescent oligonucleotide (TaqMan probe) complementary to the target sequence
  • the probe is cleaved by the 5′-3′ endonuclease activity of Taq DNA polymerase and a fluorescent dye released in the medium (Holland et al., Proc. Natl. Acad. Sci. U.S.A. 88, 7276-80, 1991). Because the fluorescence emission will increase in direct proportion to the amount of the specific amplified product, the exponential growth phase of PCR product can be detected and used to determine the initial template concentration (Heid et al., Genome Res. 6, 986-94, 1996, and Gibson et al., Genome Res. 6, 995-1001, 1996).
  • the amplification of an endogenous control can be performed to standardize the amount of sample RNA added to a reaction.
  • the control of choice is the 18S ribosomal RNA. Because reporter dyes with differing emission spectra are available, the target and the endogenous control can be independently quantified in the same tube if probes labeled with different dyes are used.
  • RNA extraction and cDNA preparation Total RNA from the tissues listed above are used for expression quantification. RNAs labeled “from autopsy” were extracted from autoptic tissues with the TRIzol reagent (Life Technologies, MD) according to the manufacturer's protocol.
  • RNA Fifty ⁇ g of each RNA were treated with DNase I for 1 hour at 37° C. in the following reaction mix: 0.2 U/ ⁇ l RNase-free DNase I (Roche Diagnostics, Germany); 0.4 U/ ⁇ l RNase inhibitor (PE Applied Biosystems, CA); 10 mM Tris-HCl pH 7.9; 10 mM MgCl 2 ; 50 mM NaCl; and 1 mM DTT.
  • RNA is extracted once with 1 volume of phenol:chloroform:—isoamyl alcohol (24:24:1) and once with chloroform, and precipitated with ⁇ fraction (1/10) ⁇ volume of 3 M NaAcetate, pH5.2, and 2 volumes of ethanol.
  • RNA from the autoptic tissues are DNase treated with the DNA-free kit purchased from Ambion (Ambion, Tex.). After resuspension and spectrophotometric quantification, each sample is reverse transcribed with the TaqMan Reverse Transcription Reagents (PE Applied Biosystems, CA) according to the manufacturer's protocol. The final concentration of RNA in the reaction mix is 200 ng/ ⁇ L. Reverse transcription is carried out with 2.5 ⁇ M of random hexamer primers.
  • TaqMan quantitative analysis Specific primers and probe are designed according to the recommendations of PE Applied Biosystems; the probe can be labeled at the 5′ end FAM (6-carboxy-fluorescein) and at the 3′ end with TAMRA (6-carboxy-tetramethyl-rhodamine). Quantification experiments are performed on 10 ng of reverse transcribed RNA from each sample. Each determination is done in triplicate.
  • FAM 6-carboxy-fluorescein
  • TAMRA 6-carboxy-tetramethyl-rhodamine
  • Total cDNA content is normalized with the simultaneous quantification (multiplex PCR) of the 18S ribosomal RNA using the Pre-Developed TaqMan Assay Reagents (PDAR) Control Kit (PE Applied Biosystems, CA).
  • PDAR Pre-Developed TaqMan Assay Reagents
  • the assay reaction mix is as follows: 1 ⁇ final TaqMan Universal PCR Master Mix (from 2 ⁇ stock) (PE Applied Biosystems, CA); 1 ⁇ PDAR control —18S RNA (from 20 ⁇ stock); 300 nM forward primer; 900 nM reverse primer; 200 nM probe; 10 ng cDNA; and water to 25 ⁇ l.
  • the cell line used for testing is the human colon cancer cell line HCT116.
  • Cells are cultured in RPMI-1640 with 10-15% fetal calf serum at a concentration of 10,000 cells per milliliter in a volume of 0.5 ml and kept at 37° C. in a 95% air/5% CO 2 atmosphere.
  • Phosphorothioate oligoribonucleotides are synthesized on an Applied Biosystems Model 380B DNA synthesizer using phosphoroamidite chemistry. A sequence of 24 bases complementary to the nucleotides at position 1 to 24 of SEQ ID NOS: 1, 8 and 10 is used as the test oligonucleotide. As a control, another (random) sequence is used: 5′-TCA ACT GAC TAG ATG TAC ATG GAC-3′. Following assembly and deprotection, oligonucleotides are ethanol-precipitated twice, dried, and suspended in phosphate buffered saline at the desired concentration.
  • oligonucleotides Purity of the oligonucleotides is tested by capillary gel electrophoresis and ion exchange HPLC. The purified oligonucleotides are added to the culture medium at a concentration of 10 ⁇ M once per day for seven days.
  • test oligonucleotide for seven days results in significantly reduced expression of human protein kinase-like protein as determined by Western blotting. This effect is not observed with the control oligonucleotide.
  • the number of cells in the cultures is counted using an automatic cell counter. The number of cells in cultures treated with the test oligonucleotide (expressed as 100%) is compared with the number of cells in cultures treated with the control oligonucleotide. The number of cells in cultures treated with the test oligonucleotide is not more than 30% of control, indicating that the inhibition of human protein kinase-like protein has an anti-proliferative effect on cancer cells.
  • This non-tumor assay measures the ability of a compound to reduce either the endogenous level of a circulating hormone or the level of hormone produced in response to a biologic stimulus.
  • Rodents are administered test compound (p.o., i.p., i.v., i.m., or s.c.).
  • test compound p.o., i.p., i.v., i.m., or s.c.
  • Plasma is assayed for levels of the hormone of interest. If the normal circulating levels of the hormone are too low and/or variable to provide consistent results, the level of the hormone may be elevated by a pre-treatment with a biologic stimulus (i.e., LHRH may be injected i.m.
  • a biologic stimulus i.e., LHRH may be injected i.m.
  • mice were fed at a dosage of 30 ng/mouse to induce a burst of testosterone synthesis).
  • the timing of plasma collection would be adjusted to coincide with the peak of the induced hormone response.
  • Compound effects are compared to a vehicle-treated control group.
  • An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test. Significance is p value ⁇ 0.05 compared to the vehicle control group.
  • Hollow fibers are prepared with desired cell line(s) and implanted intraperitoneally and/or subcutaneously in rodents. Compounds are administered p.o., i.p., i.v., i.m., or s.c. Fibers are harvested in accordance with specific readout assay protocol, these may include assays for gene expression (bDNA, PCR, or Taqman), or a specific biochemical activity (i.e., cAMP levels. Results are analyzed by Student's t-test or Rank Sum test after the variance between groups is compared by an F-test, with significance at p ⁇ 0.05 as compared to the vehicle control group.
  • specific readout assay protocol these may include assays for gene expression (bDNA, PCR, or Taqman), or a specific biochemical activity (i.e., cAMP levels. Results are analyzed by Student's t-test or Rank Sum test after the variance between groups is compared by an F-test, with significance at p ⁇ 0.05 as
  • a hormone dependent tissue i.e., seminal vesicles in males and uteri in females.
  • Rodents are administered test compound (p.o., i.p., i.v., i.m., or s.c.) according to a predetermined schedule and for
  • Organ weights may be directly compared or they may be normalized for the body weight of the animal. Compound effects are compared to a vehicle-treated control group. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test. Significance is p value ⁇ 0.05 compared to the vehicle control group.
  • Hollow fibers are prepared with desired cell line(s) and implanted intraperitoneally and/or subcutaneously in rodents. Compounds are administered p.o., i.p., i.v., i.m., or s.c. Fibers are harvested in accordance with specific readout assay protocol. Cell proliferation is determined by measuring a marker of cell number (i.e., MTT or LDH). The cell number and change in cell number from the starting inoculum are analyzed by Student's t-test or Rank Sum test after the variance between groups is compared by an F-test, with significance at p ⁇ 0.05 as compared to the vehicle control group.
  • a marker of cell number i.e., MTT or LDH
  • Hydron pellets with or without growth factors or cells are implanted into a micropocket surgically created in the rodent cornea.
  • Compound administration may be systemic or local (compound mixed with growth factors in the hydron pellet).
  • Corneas are harvested at 7 days post implantation immediately following intracardiac infusion of colloidal carbon and are fixed in 10% formalin. Readout is qualitative scoring and/or image analysis. Qualitative scores are compared by Rank Sum test. Image analysis data is evaluated by measuring the area of neovascularization (in pixels) and group averages are compared by Student's t-test (2 tail). Significance is p ⁇ 0.05 as compared to the growth factor or cells only group.
  • Matrigel containing cells or growth factors, is injected subcutaneously. Compounds are administered p.o., i.p., i.v., i.m., or s.c. Matrigel plugs are harvested at predetermined time point(s) and prepared for readout. Readout is an ELISA-based assay for hemoglobin concentration and/or histological examination (i.e. vessel count, special staining for endothelial surface markers: CD31, factor-8). Readouts are analyzed by Student's t-test, after the variance between groups is compared by an F-test, with significance determined at p ⁇ 0.05 as compared to the vehicle control group.
  • Tumor cells or fragments are implanted subcutaneously on Day 0.
  • Vehicle and/or compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule starting at a time, usually on Day 1, prior to the ability to measure the tumor burden.
  • Body weights and tumor measurements are recorded 2-3 times weekly. Mean net body and tumor weights are calculated for each data collection day.
  • Anti-tumor efficacy may be initially determined by comparing the size of treated (T) and control (C) tumors on a given day by a Student's t-test, after the variance between groups is compared by an F-test, with significance determined at p ⁇ 0.05.
  • Tumor growth delays are expressed as the difference in the median time for the treated and control groups to attain a predetermined size divided by the median time for the control group to attain that size. Growth delays are compared by generating Kaplan-Meier curves from the times for individual tumors to attain the evaluation size. Significance is p ⁇ 0.05.
  • Tumor cells are injected intraperitoneally or intracranially on Day 0.
  • Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule starting on Day 1. Observations of morbidity and/or mortality are recorded twice daily. Body weights are measured and recorded twice weekly. Morbidity/mortality data is expressed in terms of the median time of survival and the number of long-term survivors is indicated separately. Survival times are used to generate Kaplan-Meier curves. Significance is p ⁇ 0.05 by a log-rank test compared to the control group in the experiment.
  • Tumor cells or fragments are implanted subcutaneously and grown to the desired size for treatment to begin. Once at the predetermined size range, mice are randomized into treatment groups. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Tumor and body weights are measured and recorded 2-3 times weekly. Mean tumor weights of all groups over days post inoculation are graphed for comparison. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p ⁇ 0.05 as compared to the control group.
  • Tumor measurements may be recorded after dosing has stopped to monitor tumor growth delay.
  • Tumor growth delays are expressed as the difference in the median time for the treated and control groups to attain a predetermined size divided by the median time for the control group to attain that size. Growth delays are compared by generating Kaplan-Meier curves from the times for individual tumors to attain the evaluation size. Significance is p value ⁇ 0.05 compared to the vehicle control group.
  • Tumor cells or fragments, of mammary adenocarcinoma origin are implanted directly into a surgically exposed and reflected mammary fat pad in rodents. The fat pad is placed back in its original position and the surgical site is closed. Hormones may also be administered to the rodents to support the growth of the tumors. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Tumor and body weights are measured and recorded 2-3 times weekly. Mean tumor weights of all groups over days post inoculation are graphed for comparison. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p ⁇ 0.05 as compared to the control group.
  • Tumor measurements may be recorded after dosing has stopped to monitor tumor growth delay.
  • Tumor growth delays are expressed as the difference in the median time for the treated and control groups to attain a predetermined size divided by the median time for the control group to attain that size.
  • Growth delays are compared by generating Kaplan-Meier curves from the times for individual tumors to attain the evaluation size. Significance is p value ⁇ 0.05 compared to the vehicle control group.
  • this model provides an opportunity to increase the rate of spontaneous metastasis of this type of tumor. Metastasis can be assessed at termination of the study by counting the number of visible foci per target organ, or measuring the target organ weight. The means of these endpoints are compared by Student's t-test after conducting an F-test, with significance determined at p ⁇ 0.05 compared to the control group in the experiment.
  • Tumor cells or fragments, of prostatic adenocarcinoma origin are implanted directly into a surgically exposed dorsal lobe of the prostate in rodents.
  • the prostate is externalized through an abdominal incision so that the tumor can be implanted specifically in the dorsal lobe while verifying that the implant does not enter the seminal vesicles.
  • the successfully inoculated prostate is replaced in the abdomen and the incisions through the abdomen and skin are closed.
  • Hormones may also be administered to the rodents to support the growth of the tumors.
  • Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule.
  • Body weights are measured and recorded 2-3 times weekly. At a predetermined time, the experiment is terminated and the animal is dissected.
  • the size of the primary tumor is measured in three dimensions using either a caliper or an ocular micrometer attached to a dissecting scope.
  • An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p ⁇ 0.05 as compared to the control group. This model provides an opportunity to increase the rate of spontaneous metastasis of this type of tumor.
  • Metastasis can be assessed at termination of the study by counting the number of visible foci per target organ (i.e., the lungs), or measuring the target organ weight (i.e., the regional lymph nodes). The means of these endpoints are compared by Student's t-test after conducting an F-test, with significance determined at p ⁇ 0.05 compared to the control group in the experiment.
  • Tumor cells of pulmonary origin may be implanted intrabronchially by making an incision through the skin and exposing the trachea.
  • the trachea is pierced with the beveled end of a 25 gauge needle and the tumor cells are inoculated into the main bronchus using a flat-ended 27 gauge needle with a 90° bend.
  • Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Body weights are measured and recorded 2-3 times weekly. At a predetermined time, the experiment is terminated and the animal is dissected.
  • the size of the primary tumor is measured in three dimensions using either a caliper or an ocular micrometer attached to a dissecting scope.
  • An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p ⁇ 0.05 as compared to the control group.
  • This model provides an opportunity to increase the rate of spontaneous metastasis of this type of tumor. Metastasis can be assessed at termination of the study by counting the number of visible foci per target organ (i.e., the contralateral lung), or measuring the target organ weight. The means of these endpoints are compared by Student's t-test after conducting an F-test, with significance determined at p ⁇ 0.05 compared to the control group in the experiment.
  • Tumor cells of gastrointestinal origin may be implanted intracecally by making an abdominal incision through the skin and externalizing the intestine. Tumor cells are inoculated into the cecal wall without penetrating the lumen of the intestine using a 27 or 30 gauge needle. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Body weights are measured and recorded 2-3 times weekly. At a predetermined time, the experiment is terminated and the animal is dissected. The size of the primary tumor is measured in three dimensions using either a caliper or an ocular micrometer attached to a dissecting scope.
  • An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p ⁇ 0.05 as compared to the control group. This model provides an opportunity to increase the rate of spontaneous metastasis of this type of tumor. Metastasis can be assessed at termination of the study by counting the number of visible foci per target organ (i.e., the liver), or measuring the target organ weight. The means of these endpoints are compared by Student's t-test after conducting an F-test, with significance determined at p ⁇ 0.05 compared to the control group in the experiment.
  • Tumor cells are inoculated s.c. and the tumors allowed to grow to a predetermined range for spontaneous metastasis studies to the lung or liver.
  • the mean number of visible tumor foci, as determined under a dissecting microscope, and the mean target organ weights are compared by Student's t-test after conducting an F-test, with significance determined at p ⁇ 0.05 compared to the control group in the experiment for both of these endpoints.
  • Tumor cells are injected into the tail vein, portal vein, or the left ventricle of the heart in experimental (forced) lung, liver, and bone metastasis studies, respectively.
  • Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Observations of morbidity and/or mortality are recorded daily. Body weights are measured and recorded twice weekly. Potential endpoints include survival time, numbers of visible foci per target organ, or target organ weight. When survival time is used as the endpoint the other values are not determined. Survival data is used to generate Kaplan-Meier curves.
  • Significance is p ⁇ 0.05 by a log-rank test compared to the control group in the experiment.
  • the mean number of visible tumor foci, as determined under a dissecting microscope, and the mean target organ weights are compared by Student's t-test after conducting an F-test, with significance at p ⁇ 0.05 compared to the vehicle control group in the experiment for both endpoints.
  • Acute pain is measured on a hot plate mainly in rats.
  • Two variants of hot plate testing are used: In the classical variant animals are put on a hot surface (52 to 56° C.) and the latency time is measured until the animals show nocifensive behavior, such as stepping or foot licking.
  • the other variant is an increasing temperature hot plate where the experimental animals are put on a surface of neutral temperature. Subsequently this surface is slowly but constantly heated until the animals begin to lick a hind paw. The temperature which is reached when hind paw licking begins is a measure for pain threshold.
  • Compounds are tested against a vehicle treated control group. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to pain testing.
  • application routes i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal
  • Persistent pain is measured with the formalin or capsaicin test, mainly in rats.
  • a solution of 1 to 5% formalin or 10 to 100 ⁇ g capsaicin is injected into one hind paw of the experimental animal.
  • the animals show nocifensive reactions like flinching, licking and biting of the affected paw.
  • the number of nocifensive reactions within a time frame of up to 90 minutes is a measure for intensity of pain.
  • Compounds are tested against a vehicle treated control group. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to formalin or capsaicin administration.
  • application routes i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal
  • Neuropathic pain is induced by different variants of unilateral sciatic nerve injury mainly in rats.
  • the operation is performed under anesthesia.
  • the first variant of sciatic nerve injury is produced by placing loosely constrictive ligatures around the common sciatic nerve.
  • the second variant is the tight ligation of about the half of the diameter of the common sciatic nerve.
  • a group of models is used in which tight ligations or transections are made of either the L5 and L6 spinal nerves, or the L % spinal nerve only.
  • the fourth variant involves an axotomy of two of the three terminal branches of the sciatic nerve (tibial and common peroneal nerves) leaving the remaining sural nerve intact whereas the last variant comprises the axotomy of only the tibial branch leaving the sural and common nerves uninjured. Control animals are treated with a sham operation.
  • the nerve injured animals develop a chronic mechanical allodynia, cold allodynioa, as well as a thermal hyperalgesia.
  • Mechanical allodynia is measured by means of a pressure transducer (electronic von Frey Anesthesiometer, IITC Inc.-Life Science Instruments, Woodland Hills, SA, USA; Electronic von Frey System, Somedic Sales AB, Hörby, Sweden).
  • Thermal hyperalgesia is measured by means of a radiant heat source (Plantar Test, Ugo Basile, Comerio, Italy), or by means of a cold plate of 5 to 10° C. where the nocifensive reactions of the affected hind paw are counted as a measure of pain intensity.
  • a further test for cold induced pain is the counting of nocifensive reactions, or duration of nocifensive responses after plantar administration of acetone to the affected hind limb.
  • Chronic pain in general is assessed by registering the circadanian rhythms in activity (Suijo and Arndt, Universitat zu GmbH, Cologne, Germany), and by scoring differences in gait (foot print patterns; FOOTPRINTS program, Klapdor et al., 1997. A low cost method to analyze footprint patterns. J. Neurosci. Methods 75, 49-54).
  • Compounds are tested against sham operated and vehicle treated control groups. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to pain testing.
  • application routes i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal
  • Inflammatory pain is induced mainly in rats by injection of 0.75 mg carrageenan or complete Freund's adjuvant into one hind paw. The animals develop an edema with mechanical allodynia as well as thermal hyperalgesia.
  • Compounds are tested against uninflamed as well as vehicle treated control groups. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to pain testing.
  • application routes i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal
  • Compounds are tested against diabetic and non-diabetic vehicle treated control groups. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to pain testing.
  • application routes i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal
  • Degeneration of the dopaminergic nigrostriatal and striatopallidal pathways is the central pathological event in Parkinson's disease. This disorder has been mimicked experimentally in rats using single/sequential unilateral stereotaxic injections of 6-OH-DA into the medium forebrain bundle (MFB).
  • MFB medium forebrain bundle
  • Animals are administered pargyline on the day of surgery (Sigma, St. Louis, Mo., USA; 50 mg/kg i.p.) in order to inhibit metabolism of 6-OHDA by monoamine oxidase and desmethylimipramine HCl (Sigma; 25 mg/kg i.p.) in order to prevent uptake of 6-OHDA by noradrenergic terminals. Thirty minutes later the rats are anesthetized with sodium pentobarbital (50 mg/kg) and placed in a stereotaxic frame.
  • DA nigrostriatal pathway 4 ⁇ l of 0.01% ascorbic acid-saline containing 8 ⁇ g of 6-OHDA HBr (Sigma) are injected into the left medial fore-brain bundle at a rate of 1 ⁇ l/min (2.4 mm anterior, 1.49 mm lateral, ⁇ 2.7 mm ventral to Bregma and the skull surface). The needle is left in place an additional 5 min to allow diffusion to occur.
  • Forelimb akinesia is assessed three weeks following lesion placement using a modified stepping test protocol.
  • the animals are held by the experimenter with one hand fixing the hindlimbs and slightly raising the hind part above the surface.
  • One paw is touching the table, and is then moved slowly sideways (5 s for 1 m), first in the forehand and then in the backhand direction.
  • the number of adjusting steps is counted for both paws in the backhand and forehand direction of movement.
  • the sequence of testing is right paw forehand and backhand adjusting stepping, followed by left paw forehand and backhand directions.
  • the test is repeated three times on three consecutive days, after an initial training period of three days prior to the first testing.
  • Forehand adjusted stepping reveals no consistent differences between lesioned and healthy control animals. Analysis is therefore restricted to backhand adjusted stepping.
  • a modified version of the staircase test is used for evaluation of paw reaching behavior three weeks following primary and secondary lesion placement.
  • Plexiglass test boxes with a central platform and a removable staircase on each side are used.
  • the apparatus is designed such that only the paw on the same side at each staircase can be used, thus providing a measure of independent forelimb use.
  • For each test the animals are left in the test boxes for 15 min.
  • the double staircase is filled with 7 ⁇ 3 chow pellets (Precision food pellets, formula: P, purified rodent diet, size 45 mg; Sandown Scientific) on each side.
  • MPTP neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine
  • DAergic mesencephalic dopaminergic
  • TH tyrosine hydroxylase
  • mice are perfused transcardially with 0.01 M PBS (pH 7.4) for 2 min, followed by 4% paraformaldehyde (Merck) in PBS for 15 min.
  • the brains are removed and placed in 4% paraformaldehyde for 24 h at 4° C. For dehydration they are then transferred to a 20% sucrose (Merck) solution in 0.1 M PBS at 4° C. until they sink.
  • the brains are frozen in methylbutan at ⁇ 20° C. for 2 min and stored at ⁇ 70° C.
  • sledge microtome (mod. 3800-Frigocut, Leica) 25 ⁇ m sections are taken from the genu of the corpus callosum (AP 1.7 mm) to the hippocarnpus (AP 21.8 mm) and from AP 24.16 to AP 26.72. Forty-six sections are cut and stored in assorters in 0.25 M Tris buffer (pH 7.4) for immunohistochemistry.
  • TH free-floating tyrosine hydroxylase
  • the system logs the fall as the end of the experiment for that mouse, and the total time on the rotarod, as well as the time of the fall and all the set-up parameters, are recorded.
  • the system also allows a weak current to be passed through the base grid, to aid training.
  • the object recognition task has been designed to assess the effects of experimental manipulations on the cognitive performance of rodents.
  • a rat is placed in an open field, in which two identical objects are present.
  • the rats inspects both objects during the first trial of the object recognition task.
  • a second trial after a retention interval of for example 24 hours, one of the two objects used in the first trial, the ‘familiar’ object, and a novel object are placed in the open field.
  • the inspection time at each of the objects is registered.
  • the basic measures in the OR task is the time spent by a rat exploring the two object the second trial. Good retention is reflected by higher exploration times towards the novel than the ‘familiar’ object.
  • Administration of the putative cognition enhancer prior to the first trial predominantly allows assessment of the effects on acquisition, and eventually on consolidation processes.
  • Administration of the testing compound after the first trial allows to assess the effects on consolidation processes, whereas administration before the second trial allows to measure effects on retrieval processes.
  • the passive avoidance task assesses memory performance in rats and mice.
  • the inhibitory avoidance apparatus consists of a two-compartment box with a light compartment and a dark compartment. The two compartments are separated by a guillotine door that can be operated by the experimenter. A threshold of 2 cm separates the two compartments when the guillotine door is raised. When the door is open, the illumination in the dark compartment is about 2 lux. The light intensity is about 500 lux at the center of the floor of the light compartment.
  • the step-through latency that is the first latency of entering the dark compartment (in sec.) during the retention session is an index of the memory performance of the animal; the longer the latency to enter the dark compartment, the better the retention is.
  • the Morris water escape task measures spatial orientation learning in rodents. It is a test system that has extensively been used to investigate the effects of putative therapeutic on the cognitive functions of rats and mice. The performance of an animal is assessed in a circular water tank with an escape platform that is submerged about 1 cm below the surface of the water. The escape platform is not visible for an animal swimming in the water tank. Abundant extra-maze cues are provided by the furniture in the room, including desks, computer equipment, a second water tank, the presence of the experimenter, and by a radio on a shelf that is playing softly.
  • the animals receive four trials during five daily acquisition sessions.
  • a trial is started by placing an animal into the pool, facing the wall of the tank.
  • Each of four starting positions in the quadrants north, east, south, and west is used once in a series of four trials; their order is randomized.
  • the escape platform is always in the same position.
  • a trial is terminated as soon as the animal had climbs onto the escape platform or when 90 seconds have elapsed, whichever event occurs first.
  • the animal is allowed to stay on the platform for 30 seconds. Then it is taken from the platform and the next trial is started. If an animal did not find the platform within 90 seconds it is put on the platform by the experimenter and is allowed to stay there for 30 seconds.
  • an additional trial is given as a probe trial: the platform is removed, and the time the animal spends in the four quadrants is measured for 30 or 60 seconds.
  • the probe trial all animals start from the same start position, opposite to the quadrant where the escape platform had been positioned during acquisition.
  • the T-maze spontaneous alternation task assesses the spatial memory performance in mice.
  • the start arm and the two goal arms of the T-maze are provided with guillotine doors which can be operated manually by the experimenter.
  • a mouse is put into the start arm at the beginning of training.
  • the guillotine door is closed.
  • the ‘forced trial’ either the left or right goal arm is blocked by lowering the guillotine door.
  • the mouse After the mouse has been released from the start arm, it will negotiate the maze, eventually enter the open goal arm, and return to the start position, where it will be confined for 5 seconds, by lowering the guillotine door.
  • the animal can choose freely between the left and right goal arm (all guillotine-doors opened) during 14 ‘free choice’ trials. As soon a the mouse has entered one goal arm, the other one is closed. The mouse eventually returns to the start arm and is free to visit whichever go alarm it wants after having been confined to the start arm for 5 seconds. After completion of 14 free choice trials in one session, the animal is removed from the maze. During training, the animal is never handled.
  • the percent alternations out of 14 trials is calculated. This percentage and the total time needed to complete the first forced trial and the subsequent 14 free choice trials (in s) is analyzed.
  • Cognitive deficits are usually induced by an injection of scopolamine, 30 min before the start of the training session. Scopolamine reduced the per-cent alternations to chance level, or below.
  • a cognition enhancer which is always administered before the training session, will at least partially, antagonize the scopolamine-induced reduction in the spontaneous alternation rate.
  • RNA used for Taqman quantitative analysis were either purchased (Clontech,CA) or extracted from tissues using TRIzol reagent (Life Technologies, MD) according to a modified vendor protocol which utilizes the Rneasy protocol (Qiagen, CA).
  • Protein kinase-like protein expression in adipose and skeletal muscle could be regulated to increase insulin sensitivity.
  • Protein kinases 6. The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification. FASEB J 1995 May;9(8):576-96

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Reagents that regulate human protein kinase-like protein and reagents which bind to human protein kinase-like gene products can play a role in preventing, ameliorating, or correcting dysfunctions or diseases including, but not limited to, cancer, CNS disorders, COPD, obesity, diabetes, and cardiovascular disorders.

Description

  • This application incorporates by reference co-pending U.S. applications Serial No. 60/276,055 filed Mar. 16, 2001, Serial No. 60/324,530 filed Sep. 24, 2001, Serial No. 60/326,458 filed Oct. 3, 2001 and Serial No. 60/337,124 filed Dec. 10, 2001.[0001]
  • TECHNICAL FIELD OF THE INVENTION
  • The invention relates to the regulation of a human protein kinase-like protein. [0002]
  • BACKGROUND OF THE INVENTION
  • Protein kinases play critical roles in the regulation of biochemical and morphological changes associated with cellular growth and division. D'Urso et al., [0003] Science 250, 786-91, 1990; Birchmeier et al., Bioessays 15, 185-89, 1993); U.S. Pat. No. 6,200,770. They serve as growth factor receptors and signal transducers and have been implicated in cellular transformation and malignancy. Hunter et al., Cell 70, 375-87, 1992; Posada et al., Mol. Biol. Cell 3, 583-92, 1992; Hunter et al., Cell 79, 573-82, 1994. For example, protein kinases have been shown to participate in the transmission of signals from growth-factor receptors (Sturgill et al., Nature 344, 715-18, 1988; Gomez et al., Nature 353, 170-73, 1991), control of entry of cells into mitosis (Nurse et al., Nature 344, 503-08, 1990; Maller, Curr. Opin. Cell Biol. 3, 269-75, 1991), and regulation of actin bundling (Husain-Chishti et al., Nature 334, 718-21, 1988).
  • Protein kinases can be divided into two main groups based on either amino acid sequence similarity or specificity for either serine/threonine or tyrosine residues. A small number of dual-specificity kinases are structurally like the serine/threonine-specific group. Within the broad classification, kinases can be further sub-divided into families whose members share a higher degree of catalytic domain amino acid sequence identity and also have similar biochemical properties. Most protein kinase family members also share structural features outside the kinase domain that reflect their particular cellular roles. These include regulatory domains that control kinase activity or interaction with other proteins. Hanks et al., [0004] Science 241, 42-52, 1988).
  • There is a need in the art to identify protein kinase-like proteins, which can be regulated to provide therapeutic effects. [0005]
  • SUMMARY OF THE INVENTION
  • It is an object of the invention to provide reagents and methods of regulating a human protein kinase-like protein. This and other objects of the invention are provided by one or more of the embodiments described below. [0006]
  • One embodiment of the invention is a proteine kinase-like protein polypeptide comprising an amino acid sequence selected from the group consisting of: [0007]
  • amino acid sequences which are at least about 35% identical to [0008]
  • the amino acid sequence shown in SEQ ID NO: 2; [0009]
  • the amino acid sequence shown in SEQ ID NO: 2; [0010]
  • amino acid sequences which are at least about 35% identical to [0011]
  • the amino acid sequence shown in SEQ ID NO: 9 and; [0012]
  • the amino acid sequence shown in SEQ ID NO:9. [0013]
  • Yet another embodiment of the invention is a method of screening for agents which decrease extracellular matrix degradation. A test compound is contacted with a proteine kinase-like protein polypeptide comprising an amino acid sequence selected from the group consisting of: [0014]
  • amino acid sequences which are at least about 35% identical to [0015]
  • the amino acid sequence shown in SEQ ID NO: 2; [0016]
  • the amino acid sequence shown in SEQ ID NO: 2; [0017]
  • amino acid sequences which are at least about 50% identical to [0018]
  • the amino acid sequence shown in SEQ ID NO: 9 and; [0019]
  • the amino acid sequence shown in SEQ ID NO:9. [0020]
  • Binding between the test compound and the proteine kinase-like protein polypeptide is detected. A test compound which binds to the proteine kinase-like protein polypeptide is thereby identified as a potential agent for decreasing extracellular matrix degradation. The agent can work by decreasing the activity of the proteine kinase-like protein. [0021]
  • Another embodiment of the invention is a method of screening for agents which decrease extracellular matrix degradation. A test compound is contacted with a polynucleotide encoding a proteine kinase-like protein polypeptide, wherein the polynucleotide comprises a nucleotide sequence selected from the group consisting of: [0022]
  • nucleotide sequences which are at least about 50% identical to [0023]
  • the nucleotide sequence shown in SEQ ID NO: 1; [0024]
  • the nucleotide sequence shown in SEQ ID NO: 1; [0025]
  • nucleotide sequences which are at least about 50% identical to [0026]
  • the nucleotide sequence shown in SEQ ID NO: 8; [0027]
  • the nucleotide sequence shown in SEQ ID NO: 8; [0028]
  • nucleotide sequences which are at least about 50% identical to [0029]
  • the nucleotide sequence shown in SEQ ID NO: 10; and [0030]
  • the nucleotide sequence shown in SEQ ID NO: 10. [0031]
  • Binding of the test compound to the polynucleotide is detected. A test compound which binds to the polynucleotide is identified as a potential agent for decreasing extracellular matrix degradation. The agent can work by decreasing the amount of the proteine kinase-like protein through interacting with the proteine kinase-like protein mRNA. [0032]
  • Another embodiment of the invention is a method of screening for agents which regulate extracellular matrix degradation. A test compound is contacted with a proteine kinase-like protein polypeptide comprising an amino acid sequence selected from the group consisting of: [0033]
  • amino acid sequences which are at least about 35% identical to [0034]
  • the amino acid sequence shown in SEQ ID NO: 2; [0035]
  • the amino acid sequence shown in SEQ ID NO: 2; [0036]
  • amino acid sequences which are at least about 35% identical to [0037]
  • the amino acid sequence shown in SEQ ID NO: 9 and; [0038]
  • the amino acid sequence shown in SEQ ID NO:9. [0039]
  • A proteine kinase-like protein activity of the polypeptide is detected. A test compound which increases proteine kinase-like protein activity of the polypeptide relative to proteine kinase-like protein activity in the absence of the test compound is thereby identified as a potential agent for increasing extracellular matrix degradation. A test compound which decreases proteine kinase-like protein activity of the polypeptide relative to proteine kinase-like protein activity in the absence of the test compound is thereby identified as a potential agent for decreasing extracellular matrix degradation. [0040]
  • Even another embodiment of the invention is a method of screening for agents which decrease extracellular matrix degradation. A test compound is contacted with a proteine kinase-like protein product of a polynucleotide which comprises a nucleotide sequence selected from the group consisting of: [0041]
  • nucleotide sequences which are at least about 50% identical to [0042]
  • the nucleotide sequence shown in SEQ ID NO: 1; [0043]
  • the nucleotide sequence shown in SEQ ID NO: 1; [0044]
  • nucleotide sequences which are at least about 50% identical to [0045]
  • the nucleotide sequence shown in SEQ ID NO: 8; [0046]
  • the nucleotide sequence shown in SEQ ID NO: 8; [0047]
  • nucleotide sequences which are at least about 50% identical to [0048]
  • the nucleotide sequence shown in SEQ ID NO: 10; and [0049]
  • the nucleotide sequence shown in SEQ ID NO:10. [0050]
  • Binding of the test compound to the proteine kinase-like protein product is detected. A test compound which binds to the proteine kinase-like protein product is thereby identified as a potential agent for decreasing extracellular matrix degradation. [0051]
  • Still another embodiment of the invention is a method of reducing extracellular matrix degradation. A cell is contacted with a reagent which specifically binds to a polynucleotide encoding a proteine kinase-like protein polypeptide or the product encoded by the polynucleotide, wherein the polynucleotide comprises a nucleotide sequence selected from the group consisting of: [0052]
  • nucleotide sequences which are at least about 50% identical to [0053]
  • the nucleotide sequence shown in SEQ ID NO: 1; [0054]
  • the nucleotide sequence shown in SEQ ID NO: 1; [0055]
  • nucleotide sequences which are at least about 50% identical to [0056]
  • the nucleotide sequence shown in SEQ ID NO: 8; [0057]
  • the nucleotide sequence shown in SEQ ID NO: 8; [0058]
  • nucleotide sequences which are at least about 50% identical to [0059]
  • the nucleotide sequence shown in SEQ ID NO: 10; and [0060]
  • the nucleotide sequence shown in SEQ ID NO: 10. [0061]
  • Proteine kinase-like protein activity in the cell is thereby decreased. [0062]
  • The invention thus provides a human protein kinase-like protein that can be used to identify test compounds that may act, for example, as activators or inhibitors at the enzyme's active site. Human protein kinase-like protein and fragments thereof also are useful in raising specific antibodies that can block the enzyme and effectively reduce its activity.[0063]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:1). [0064]
  • FIG. 2 shows the amino acid sequence deduced from the DNA-sequence of FIG. 1 (SEQ ID NO:2). [0065]
  • FIG. 3 shows the amino acid sequence of the protein identified by swiss|P80192∥M3K9_HUMAN (SEQ ID NO:3). [0066]
  • FIG. 4 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:4). [0067]
  • FIG. 5 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:5). [0068]
  • FIG. 6 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:6). [0069]
  • FIG. 7 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:7). [0070]
  • FIG. 8 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:8). [0071]
  • FIG. 9 shows the amino acid sequence deduced from the DNA-sequence of FIG. 8 (SEQ ID NO:9). [0072]
  • FIG. 10 shows the DNA-sequence encoding a proteine kinase-like protein Polypeptide (SEQ ID NO:10). [0073]
  • FIG. 11 shows the BLASTP-alignment of 423 (SEQ ID NO:2) against swiss|P801921M3K9-HUMAN (SEQ ID NO:3). [0074]
  • FIG. 12 shows the HMMPFAM—alignment of 423 (SEQ ID NO:2) against pfam|hmm|pkinase. [0075]
  • FIG. 13 shows the relative expression of human protein kinase-like protein mRNA in cancer cells and tissue. [0076]
  • FIG. 14 shows the relative expression of protein kinase-like protein mRNA in tissue relevant for obesity and diabetes. [0077]
  • FIG. 15 shows the copy number of protein kinase-like protein mRNA in tissue relevant for obesity and diabetes. [0078]
  • FIG. 16 shows the relative expression profile of protein kinase-like protein mRNA in various human tissues (bodymap).[0079]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention relates to an isolated polynucleotide from the group consisting of: [0080]
  • a) a polynucleotide encoding a proteine kinase-like protein polypeptide comprising an amino acid sequence selected from the group consisting of: [0081]
  • amino acid sequences which are at least about 35% identical to the amino acid sequence shown in SEQ ID NO: 2; [0082]
  • the amino acid sequence shown in SEQ ID NO: 2; [0083]
  • amino acid sequences which are at least about 35% identical to the amino acid sequence shown in SEQ ID NO: 9 and; [0084]
  • the amino acid sequence shown in SEQ ID NO:9. [0085]
  • b) a polynucleotide comprising the sequence of SEQ ID NOS: 1, 8 or 10; [0086]
  • c) a polynucleotide which hybridizes under stringent conditions to a polynucleotide specified in (a) and (b) and encodes a proteine kinase-like protein polypeptide; [0087]
  • d) a polynucleotide the sequence of which deviates from the polynucleotide sequences specified in (a) to (c) due to the degeneration of the genetic code and encodes a proteine kinase-like protein polypeptide; and [0088]
  • e) a polynucleotide which represents a fragment, derivative or allelic variation of a polynucleotide sequence specified in (a) to (d) and encodes a proteine kinase-like protein polypeptide. [0089]
  • Furthermore, it has been discovered by the present applicant that a novel protein kinase-like protein, particularly a human protein kinase-like protein, can be used in therapeutic methods to treat cancer, a CNS disorder, COPD, obesity, diabetes, or a cardiovascular disorder. [0090]
  • Human protein kinase-like protein comprises the amino acid sequence shown in SEQ ID NOS:2 and 9. A coding sequence for human protein kinase-like protein is shown in SEQ ID NOS: 1, 8 and 10. This sequence is located on chromosome 15. Related ESTs (SEQ ID NOS:4-7) are expressed in lymphoma and germinal center B cells. [0091]
  • Human protein kinase-like protein is 31% identical over 287 amino acids to swiss|P80192|M3K9_HUMAN (SEQ ID NO:3) (FIG. 1). Pfam homology search results support the identification of this protein as a eukaryotic protein kinase. Its likely function as a tyrosine kinase class m protein also is supported by the results of a BLOCKS database search. Consensus patterns are underlined in FIG. 1; the active site Asp residue and the ATP-binding Lys residue are highlighted in bold. [0092]
  • Human protein kinase-like protein of the invention is expected to be useful for the same purposes as previously identified protein kinase-like protein enzymes. Human protein kinase-like protein is believed to be useful in therapeutic methods to treat disorders such as cancer, CNS disorders, COPD, obesity, diabetes, and cardiovascular disorders. Human protein kinase-like protein also can be used to screen for human protein kinase-like protein activators and inhibitors. [0093]
  • Polypeptides [0094]
  • Human protein kinase-like polypeptides according to the invention comprise at least 6, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, or 286 contiguous amino acids selected from the amino acid sequence shown in SEQ ID NO:2 or a biologically active variant thereof, as defined below. Human protein kinase-like polypeptides according to the invention comprise at least 6, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, 700, 725, 750, 775, 800, 825, 850, 875, 900, 925, 950, 975, 1000, 1025, 1050, 1075, 1100, 1125, 1150, 1175, 1200, 1225, 1250, 1275, 1300, 1325, 1350, 1375 or 1394 contiguous amino acids selected from the amino acid sequence shown in SEQ ID NO:9 or a biologically active variant thereof, as defined below. A protein kinase-like polypeptide of the invention therefore can be a portion of a protein kinase-like protein, a full-length protein kinase-like protein, or a fusion protein comprising all or a portion of a protein kinase-like protein. [0095]
  • Biologically Active Variants [0096]
  • Human protein kinase-like polypeptide variants which are biologically active, e.g., retain enzymatic activity, also are human protein kinase-like polypeptides. Preferably, naturally or non-naturally occurring human protein kinase-like polypeptide variants have amino acid sequences which are at least about 32, 35, 40, 45, 50, 55, 60, 65, or 70, preferably about 75, 80, 85, 90, 96, 96, 98, or 99% identical to the amino acid sequence shown in SEQ ID NOS:2 and 9 or a fragment thereof. Percent identity between a putative human protein kinase-like polypeptide variant and an amino acid sequence of SEQ ID NO:2 or 9 is determined by conventional methods. See, for example, Altschul et al., [0097] Bull. Math. Bio. 48:603 (1986), and Henikoff & Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1992). Briefly, two amino acid sequences are aligned to optimize the alignment scores using a gap opening penalty of 10, a gap extension penalty of 1, and the “BLOSUM62” scoring matrix of Henikoff & Henikoff, 1992.
  • Those skilled in the art appreciate that there are many established algorithms available to align two amino acid sequences. The “FASTA” similarity search algorithm of Pearson & Lipman is a suitable protein alignment method for examining the level of identity shared by an amino acid sequence disclosed herein and the amino acid sequence of a putative variant. The FASTA algorithm is described by Pearson & Lipman, [0098] Proc. Nat'l Acad. Sci. USA 85:2444(1988), and by Pearson, Meth. Enzymol. 183:63 (1990). Briefly, FASTA first characterizes sequence similarity by identifying regions shared by the query sequence (e.g., SEQ ID NO: 2 or 9) and a test sequence that have either the highest density of identities (if the ktup variable is 1) or pairs of identities (if ktup=2), without considering conservative amino acid substitutions, insertions, or deletions. The ten regions with the highest density of identities are then rescored by comparing the similarity of all paired amino acids using an amino acid substitution matrix, and the ends of the regions are “trimmed” to include only those residues that contribute to the highest score. If there are several regions with scores greater than the “cutoff” value (calculated by a predetermined formula based upon the length of the sequence the ktup value), then the trimmed initial regions are examined to determine whether the regions can be joined to form an approximate alignment with gaps. Finally, the highest scoring regions of the two amino acid sequences are aligned using a modification of the Needleman-Wunsch-Sellers algorithm (Needleman & Wunsch, J. Mol. Biol.48:444 (1970); Sellers, SIAM J. Appl. Math.26:787 (1974)), which allows for amino acid insertions and deletions. Preferred parameters for FASTA analysis are: ktup=1, gap opening penalty=10, gap extension penalty=1, and substitution matrix=BLOSUM62. These parameters can be introduced into a FASTA program by modifying the scoring matrix file (“SMATRIX”), as explained in Appendix 2 of Pearson, Meth. Enzymol. 183:63 (1990).
  • FASTA can also be used to determine the sequence identity of nucleic acid molecules using a ratio as disclosed above. For nucleotide sequence comparisons, the ktup value can range between one to six, preferably from three to six, most preferably three, with other parameters set as default. [0099]
  • Variations in percent identity can be due, for example, to amino acid substitutions, insertions, or deletions. Amino acid substitutions are defined as one for one amino acid replacements. They are conservative in nature when the substituted amino acid has similar structural and/or chemical properties. Examples of conservative replacements are substitution of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine. [0100]
  • Amino acid insertions or deletions are changes to or within an amino acid sequence. They typically fall in the range of about 1 to 5 amino acids. Guidance in determining which amino acid residues can be substituted, inserted, or deleted without abolishing biological or immunological activity of a human protein kinase-like polypeptide can be found using computer programs well known in the art, such as DNASTAR software. [0101]
  • The invention additionally, encompasses protein kinase-like polypeptides that are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications can be carried out by known techniques including, but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH[0102] 4, acetylation, formylation, oxidation, reduction, metabolic synthesis in the presence of tunicamycin, etc.
  • Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of prokaryotic host cell expression. The protein kinase-like polypeptides may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein. [0103]
  • The invention also provides chemically modified derivatives of protein kinase-like polypeptides that may provide additional advantages such as increased solubility, stability and circulating time of the polypeptide, or decreased immunogenicity (see U.S. Pat. No. 4,179,337). The chemical moieties for derivitization can be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol, and the like. The polypeptides can be modified at random or predetermined positions within the molecule and can include one, two, three, or more attached chemical moieties. [0104]
  • Whether an amino acid change or a polypeptide modification results in a biologically active protein kinase-like polypeptide can readily be determined by assaying for enzymatic activity, as described for example, in U.S. Pat. No. 6,194,186. [0105]
  • Fusion Proteins [0106]
  • Fusion proteins are useful for generating antibodies against protein kinase-like polypeptide amino acid sequences and for use in various assay systems. For example, fusion proteins can be used to identify proteins that interact with portions of a protein kinase-like polypeptide. Protein affinity chromatography or library-based assays for protein-protein interactions, such as the yeast two-hybrid or phage display systems, can be used for this purpose. Such methods are well known in the art and also can be used as drug screens. [0107]
  • A protein kinase-like polypeptide fusion protein comprises two polypeptide segments fused together by means of a peptide bond. The first polypeptide segment comprises at least 6, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, or 286 contiguous amino acids of SEQ D NOS:2 or of a biologically active variant, such as those described above. The first polypeptide segment comprises at least 6, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, 700, 725, 750, 775, 800, 825, 850, 875, 900, 925, 950, 975, 1000, 1025, 1050, 1075, 1100, 1125, 1150, 1175, 1200, 1225, 1250, 1275, 1300, 1325, 1350, 1375 or 1394 contiguous amino acids of SEQ ID NOS:2 or of a biologically active variant, such as those described above. The first polypeptide segment also can comprise full-length protein kinase-like protein. [0108]
  • The second polypeptide segment can be a full-length protein or a protein fragment. Proteins commonly used in fusion protein construction include 13-galactosidase, P-glucuronidase, green fluorescent protein (GFP), autofluorescent proteins, including blue fluorescent protein (BFP), glutathione-S-transferase (GST), luciferase, horseradish peroxidase (HRP), and chloramphenicol acetyltransferase (CAT). Additionally, epitope tags are used in fusion protein constructions, including histidine (His) tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, VSV-G tags, and thioredoxin (Trx) tags. Other fusion constructions can include maltose binding protein (MBP), S-tag, Lex a DNA binding domain (DBD) fusions, GALA DNA binding domain fusions, and herpes simplex virus (HSV) BP16 protein fusions. A fusion protein also can be engineered to contain a cleavage site located between the protein kinase-like polypeptide-encoding sequence and the heterologous protein sequence, so that the protein kinase-like polypeptide can be cleaved and purified away from the heterologous moiety. [0109]
  • A fusion protein can be synthesized chemically, as is known in the art. Preferably, a fusion protein is produced by covalently linking two polypeptide segments or by standard procedures in the art of molecular biology. Recombinant DNA methods can be used to prepare fusion proteins, for example, by making a DNA construct which comprises coding sequences selected from SEQ ID NOS: 1, 8 and 10 in proper reading frame with nucleotides encoding the second polypeptide segment and expressing the DNA construct in a host cell, as is known in the art. Many kits for constructing fusion proteins are available from companies such as Promega Corporation (Madison, Wis.), Stratagene (La Jolla, Calif.), CLONTECH (Mountain View, Calif.), Santa Cruz Biotechnology (Santa Cruz, Calif.), MBL International Corporation (MIC; Watertown, Mass.), and Quantum Biotechnologies (Montreal, Canada; 1-888-DNA-KITS). [0110]
  • Identification of Species Homologs [0111]
  • Species homologs of human protein kinase-like polypeptide can be obtained using protein kinase-like polypeptide polynucleotides (described below) to make suitable probes or primers for screening cDNA expression libraries from other species, such as mice, monkeys, or yeast, identifying cDNAs which encode homologs of protein kinase-like polypeptide, and expressing the cDNAs as is known in the art. [0112]
  • Polynucleotides [0113]
  • A protein kinase-like polynucleotide can be single- or double-stranded and comprises a coding sequence or the complement of a coding sequence for a protein kinase-like polypeptide. A coding sequence for human protein kinase-like protein is shown in SEQ ID NOS: 1, 8 and 10. [0114]
  • Degenerate nucleotide sequences encoding human protein kinase-like polypeptides, as well as homologous nucleotide sequences which are at least about 50, 55, 60, 65, 70, preferably about 75, 90, 96, 98, or 99% identical to the nucleotide sequence shown in SEQ ID NOS: 1, 8 and 10 or its complement also are protein kinase-like polynucleotides. Percent sequence identity between the sequences of two polynucleotides is determined using computer programs such as ALIGN which employ the FASTA algorithm, using an affine gap search with a gap open penalty of −12 and a gap extension penalty of −2. Complementary DNA (cDNA) molecules, species homologs, and variants of protein kinase-like polynucleotides that encode biologically active protein kinase-like polypeptides also are protein kinase-like polynucleotides. Polynucleotide fragments comprising at least 8, 9, 10, 11, 12, 15, 20, or 25 contiguous nucleotides of SEQ ID NOS: 1, 8 and 10 or its complement also are protein kinase-like polynucleotides. These fragments can be used, for example, as hybridization probes or as antisense oligonucleotides. [0115]
  • Identification of Polynucleotide Variants and Homologs [0116]
  • Variants and homologs of the protein kinase-like polynucleotides described above also are protein kinase-like polynucleotides. Typically, homologous protein kinase-like polynucleotide sequences can be identified by hybridization of candidate polynucleotides to known protein kinase-like polynucleotides under stringent conditions, as is known in the art. For example, using the following wash conditions—2×SSC (0.3 M NaCl, 0.03 M sodium citrate, pH 7.0), 0.1% SDS, room temperature twice, 30 minutes each; then 2×SSC, 0.1% SDS, 50° C. once, 30 minutes; then 2×SSC, room temperature twice, 10 minutes each—homologous sequences can be identified which contain at most about 25-30% basepair mismatches. More preferably, homologous nucleic acid strands contain 15-25% basepair mismatches, even more preferably 5-15% basepair mismatches. [0117]
  • Species homologs of the protein kinase-like polynucleotides disclosed herein also can be identified by making suitable probes or primers and screening cDNA expression libraries from other species, such as mice, monkeys, or yeast. Human variants of protein kinase-like polynucleotides can be identified, for example, by screening human cDNA expression libraries. It is well known that the T[0118] m of a double-stranded DNA decreases by 1-1.5° C. with every 1% decrease in homology (Bonner et al., J.
  • Mol. Biol. 81, 123 (1973). Variants of human protein kinase-like polynucleotides or protein kinase-like polynucleotides of other species can therefore be identified by hybridizing a putative homologous protein kinase-like polynucleotide with a polynucleotide having a nucleotide sequence of SEQ ID NOS: 1, 8 and 10 or the complement thereof to form a test hybrid. The melting temperature of the test hybrid is compared with the melting temperature of a hybrid comprising polynucleotides having perfectly complementary nucleotide sequences, and the number or percent of basepair mismatches within the test hybrid is calculated. [0119]
  • Nucleotide sequences which hybridize to protein kinase-like polynucleotides or their complements following stringent hybridization and/or wash conditions also are protein kinase-like polynucleotides. Stringent wash conditions are well known and understood in the art and are disclosed, for example, in Sambrook et al., M[0120] OLECULAR CLONING: A LABORATORY MANUAL, 2d ed., 1989, at pages 9.50-9.51.
  • Typically, for stringent hybridization conditions a combination of temperature and salt concentration should be chosen that is approximately 12-20° C. below the calculated T[0121] m of the hybrid under study. The Tm of a hybrid between a protein kinase-like polynucleotide having a nucleotide sequence shown in SEQ ID NOS: 1, 8 and 10 or the complement thereof and a polynucleotide sequence which is at least about 50, preferably about 75, 90, 96, or 98% identical to one of those nucleotide sequences can be calculated, for example, using the equation of Bolton and McCarthy, Proc. Natl. Acad. Sci. U.S.A. 48, 1390 (1962):
  • T m=81.5° C.-16.6(log10[Na+])+0.41(% G+C)-0.63(% formamide)-600/l),
  • where l=the length of the hybrid in basepairs. [0122]
  • Stringent wash conditions include, for example, 4×SSC at 65° C., or 50% formamide, 4×SSC at 42° C., or 0.5×SSC, 0.1% SDS at 65° C. Highly stringent wash conditions include, for example, 0.2×SSC at 65° C. [0123]
  • Preparation of Polynucleotides [0124]
  • A protein kinase-like polynucleotide can be isolated free of other cellular components such as membrane components, proteins, and lipids. Polynucleotides can be made by a cell and isolated using standard nucleic acid purification techniques, or synthesized using an amplification technique, such as the polymerase chain reaction (PCR), or by using an automatic synthesizer. Methods for isolating polynucleotides are routine and are known in the art. Any such technique for obtaining a polynucleotide can be used to obtain isolated protein kinase-like polynucleotides. For example, restriction enzymes and probes can be used to isolate polynucleotide fragments, which comprise protein kinase-like protein nucleotide sequences. Isolated polynucleotides are in preparations that are free or at least 70, 80, or 90% free of other molecules. [0125]
  • Human protein kinase-like cDNA molecules can be made with standard molecular biology techniques, using protein kinase-like mRNA as a template. Human protein kinase-like cDNA molecules can thereafter be replicated using molecular biology techniques known in the art and disclosed in manuals such as Sambrook et al. (1989). An amplification technique, such as PCR, can be used to obtain additional copies of polynucleotides of the invention, using either human genomic DNA or cDNA as a template. [0126]
  • Alternatively, synthetic chemistry techniques can be used to synthesize protein kinase-like polynucleotides. The degeneracy of the genetic code allows alternate nucleotide sequences to be synthesized which will encode a protein kinase-like polypeptide having, for example, an amino acid sequence shown in SEQ ID NOS:2 and 9 or a biologically active variant thereof. [0127]
  • Extending Polynucleotides [0128]
  • The partial sequence disclosed herein can be used to identify the corresponding full length gene from which it was derived. The partial sequence can be nick-translated or end-labeled with [0129] 32P using polynucleotide kinase using labeling methods known to those with skill in the art (BASIC METHODS IN MOLECULAR BIOLOGY, Davis et al., eds., Elsevier Press, N.Y., 1986). A lambda library prepared from human tissue can be directly screened with the labeled sequences of interest or the library can be converted en masse to pBluescript (Stratagene Cloning Systems, La Jolla, Calif. 92037) to facilitate bacterial colony screening (see Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, Cold Spring Harbor Laboratory Press (1989, pg. 1.20).
  • Both methods are well known in the art. Briefly, filters with bacterial colonies containing the library in pBluescript or bacterial lawns containing lambda plaques are denatured, and the DNA is fixed to the filters. The filters are hybridized with the labeled probe using hybridization conditions described by Davis et al., 1986. The partial sequences, cloned into lambda or pBluescript, can be used as positive controls to assess background binding and to adjust the hybridization and washing stringencies necessary for accurate clone identification. The resulting autoradiograms are compared to duplicate plates of colonies or plaques; each exposed spot corresponds to a positive colony or plaque. The colonies or plaques are selected, expanded and the DNA is isolated from the colonies for further analysis and sequencing. [0130]
  • Positive cDNA clones are analyzed to determine the amount of additional sequence they contain using PCR with one primer from the partial sequence and the other primer from the vector. Clones with a larger vector-insert PCR product than the original partial sequence are analyzed by restriction digestion and DNA sequencing to determine whether they contain an insert of the same size or similar as the mRNA size determined from Northern blot Analysis. [0131]
  • Once one or more overlapping cDNA clones are identified, the complete sequence of the clones can be determined, for example after exonuclease III digestion (McCombie et al., [0132] Methods 3, 33-40, 1991). A series of deletion clones are generated, each of which is sequenced. The resulting overlapping sequences are assembled into a single contiguous sequence of high redundancy (usually three to five overlapping sequences at each nucleotide position), resulting in a highly accurate final sequence.
  • Various PCR-based methods can be used to extend the nucleic acid sequences disclosed herein to detect upstream sequences such as promoters and regulatory elements. For example, restriction-site PCR uses universal primers to retrieve unknown sequence adjacent to a known locus (Sarkar, [0133] PCR Methods Applic. 2, 318-322, 1993). Genomic DNA is first amplified in the presence of a primer to a linker sequence and a primer specific to the known region. The amplified sequences are then subjected to a second round of PCR with the same linker primer and another specific primer internal to the first one. Products of each round of PCR are transcribed with an appropriate RNA polymerase and sequenced using reverse transcriptase.
  • Inverse PCR also can be used to amplify or extend sequences using divergent primers based on a known region (Triglia et al., [0134] Nucleic Acids Res. 16, 8186, 1988). Primers can be designed using commercially available software, such as OLIGO 4.06 Primer Analysis software (National Biosciences Inc., Plymouth, Minn.), to be 22-30 nucleotides in length, to have a GC content of 50% or more, and to anneal to the target sequence at temperatures about 68-72° C. The method uses several restriction enzymes to generate a suitable fragment in the known region of a gene. The fragment is then circularized by intramolecular ligation and used as a PCR template.
  • Another method which can be used is capture PCR, which involves PCR amplification of DNA fragments adjacent to a known sequence in human and yeast artificial chromosome DNA (Lagerstrom et al., [0135] PCR Methods Applic. 1, 111-119, 1991). In this method, multiple restriction enzyme digestions and ligations also can be used to place an engineered double-stranded sequence into an unknown fragment of the DNA molecule before performing PCR.
  • Another method which can be used to retrieve unknown sequences is that of Parker et al., [0136] Nucleic Acids Res. 19, 3055-3060, 1991). Additionally, PCR, nested primers, and PROMOTERFINDER libraries (CLONTECH, Palo Alto, Calif) can be used to walk genomic DNA (CLONTECH, Palo Alto, Calif.). This process avoids the need to screen libraries and is useful in finding intron/exon junctions.
  • When screening for full-length cDNAs, it is preferable to use libraries that have been size-selected to include larger cDNAs. Randomly-primed libraries are preferable, in that they will contain more sequences which contain the 5′ regions of genes. Use of a randomly primed library may be especially preferable for situations in which an oligo d(T) library does not yield a full-length cDNA. Genomic libraries can be useful for extension of sequence into 5′ non-transcribed regulatory regions. [0137]
  • Commercially available capillary electrophoresis systems can be used to analyze the size or confirm the nucleotide sequence of PCR or sequencing products. For example, capillary sequencing can employ flowable polymers for electrophoretic separation, four different fluorescent dyes (one for each nucleotide) that are laser activated, and detection of the emitted wavelengths by a charge coupled device camera. Output/light intensity can be converted to electrical signal using appropriate software (e.g. GENOTYPER and Sequence NAVIGATOR, Perkin Elmer), and the entire process from loading of samples to computer analysis and electronic data display can be computer controlled. Capillary electrophoresis is especially preferable for the sequencing of small pieces of DNA that might be present in limited amounts in a particular sample. [0138]
  • Obtaining Polypeptides [0139]
  • Human protein kinase-like polypeptides can be obtained, for example, by purification from human or other cells, by expression of protein kinase-like polynucleotides, or by direct chemical synthesis. [0140]
  • Protein Purification [0141]
  • Human protein kinase-like polypeptides can be purified from any cell that expresses the polypeptide, including host cells that have been transfected with protein kinase-like protein expression constructs. A purified protein kinase-like polypeptide is separated from other compounds that normally associate with the protein kinase-like polypeptide in the cell, such as certain proteins, carbohydrates, or lipids, using methods well-known in the art. Such methods include, but are not limited to, size exclusion chromatography, ammonium sulfate fractionation, ion exchange chromatography, affinity chromatography, and preparative gel electrophoresis. A preparation of purified protein kinase-like polypeptides is at least 80% pure; preferably, the preparations are 90%, 95%, or 99% pure. Purity of the preparations can be assessed by any means known in the art, such as SDS-polyacrylamide gel electrophoresis. [0142]
  • Expression of Polynucleotides [0143]
  • To express a protein kinase-like polynucleotide, the polynucleotide can be inserted into an expression vector that contains the necessary elements for the transcription and translation of the inserted coding sequence. Methods that are well known to those skilled in the art can be used to construct expression vectors containing sequences encoding protein kinase-like polypeptides and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described, for example, in Sambrook et al. (1989) and in Ausubel et al., C[0144] URRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, N.Y., 1989.
  • A variety of expression vector/host systems can be utilized to contain and express sequences encoding a protein kinase-like polypeptide. These include, but are not limited to, microorganisms, such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors, insect cell systems infected with virus expression vectors (e.g., baculovirus), plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids), or animal cell systems. [0145]
  • The control elements or regulatory sequences are those non-translated regions of the vector—enhancers, promoters, 5′ and 3′ untranslated regions—which interact with host cellular proteins to carry out transcription and translation. Such elements can vary in their strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, can be used. For example, when cloning in bacterial systems, inducible promoters such as the hybrid lacZ promoter of the BLUESCRIPT phagemid (Stratagene, LaJolla, Calif.) or pSPORT1 plasmid (Life Technologies) and the like can be used. The baculovirus polyhedrin promoter can be used in insect cells. Promoters or enhancers derived from the genomes of plant cells (e.g., heat shock, RUBISCO, and storage genes) or from plant viruses (e.g., viral promoters or leader sequences) can be cloned into the vector. In mammalian cell systems, promoters from mammalian genes or from mammalian viruses are preferable. If it is necessary to generate a cell line that contains multiple copies of a nucleotide sequence encoding a protein kinase-like polypeptide, vectors based on SV40 or EBV can be used with an appropriate selectable marker. [0146]
  • Bacterial and Yeast Expression Systems [0147]
  • In bacterial systems, a number of expression vectors can be selected depending upon the use intended for the protein kinase-like polypeptide. For example, when a large quantity of a protein kinase-like polypeptide is needed for the induction of antibodies, vectors which direct high level expression of fusion proteins that are readily purified can be used. Such vectors include, but are not limited to, multifunctional [0148] E. coli cloning and expression vectors such as BLUESCRIPT (Stratagene). In a BLUESCRIPT vector, a sequence encoding the protein kinase-like polypeptide can be ligated into the vector in frame with sequences for the amino-terminal Met and the subsequent 7 residues of β-galactosidase so that a hybrid protein is produced. pIN vectors (Van Heeke & Schuster, J. Biol. Chem. 264, 5503-5509, 1989) or pGEX vectors (Promega, Madison, Wis.) also can be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. Proteins made in such systems can be designed to include heparin, thrombin, or factor Xa protease cleavage sites so that the cloned polypeptide of interest can be released from the GST moiety at will.
  • In the yeast [0149] Saccharomyces cerevisiae, a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH can be used. For reviews, see Ausubel et al. (1989) and Grant et al., Methods Enzymol. 153, 516-544, 1987.
  • Plant and Insect Expression Systems [0150]
  • If plant expression vectors are used, the expression of sequences encoding protein kinase-like polypeptides can be driven by any of a number of promoters. For example, viral promoters such as the 35S and 19S promoters of CaMV can be used alone or in combination with the omega leader sequence from TMV (Takamatsu, [0151] EMBO J. 6, 307-311, 1987). Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters can be used (Coruzzi et al., EMBO J. 3, 1671-1680, 1984; Broglie et al., Science 224, 838-843, 1984; Winter et al., Results Probl. Cell Differ. 17, 85-105, 1991). These constructs can be introduced into plant cells by direct DNA transformation or by pathogen-mediated transfection. Such techniques are described in a number of generally available reviews (e.g., Hobbs or Murray, in MCGRAW HILL YEARBOOK OF SCIENCE AND TECHNOLOGY, McGraw Hill, New York, N.Y., pp. 191-196,1992).
  • An insect system also can be used to express a protein kinase-like polypeptide. For example, in one such system [0152] Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae. Sequences encoding protein kinase-like polypeptides can be cloned into a non-essential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of protein kinase-like polypeptides will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein. The recombinant viruses can then be used to infect S. frugiperda cells or Trichoplusia larvae in which protein kinase-like polypeptides can be expressed (Engelhard et al., Proc. Nat. Acad. Sci. 91, 3224-3227, 1994).
  • Mammalian Expression Systems [0153]
  • A number of viral-based expression systems can be used to express protein kinase-like polypeptides in mammalian host cells. For example, if an adenovirus is used as an expression vector, sequences encoding protein kinase-like polypeptides can be ligated into an adenovirus transcription/translation complex comprising the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome can be used to obtain a viable virus that is capable of expressing a protein kinase-like polypeptide in infected host cells (Logan & Shenk, [0154] Proc. Natl. Acad. Sci. 81, 3655-3659, 1984). If desired, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, can be used to increase expression in mammalian host cells.
  • Human artificial chromosomes (HACs) also can be used to deliver larger fragments of DNA than can be contained and expressed in a plasmid. HACs of 6M to 10M are constructed and delivered to cells via conventional delivery methods (e.g., liposomes, polycationic amino polymers, or vesicles). [0155]
  • Specific initiation signals also can be used to achieve more efficient translation of sequences encoding protein kinase-like polypeptides. Such signals include the ATG initiation codon and adjacent sequences. In cases where sequences encoding a protein kinase-like polypeptide, its initiation codon, and upstream sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a fragment thereof, is inserted, exogenous translational control signals (including the ATG initiation codon) should be provided. The initiation codon should be in the correct reading frame to ensure translation of the entire insert. Exogenous translational elements and initiation codons can be of various origins, both natural and synthetic. The efficiency of expression can be enhanced by the inclusion of enhancers which are appropriate for the particular cell system which is used (see Scharf et al., [0156] Results Probl. Cell Differ. 20, 125-162, 1994).
  • Host Cells [0157]
  • A host cell strain can be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein kinase-like polypeptide in the desired fashion. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation. Post-translational processing which cleaves a “prepro” form of the polypeptide also can be used to facilitate correct insertion, folding and/or function. Different host cells that have specific cellular machinery and characteristic mechanisms for post-translational activities (e.g., CHO, HeLa, MDCK, HEK293, and WI38), are available from the American Type Culture Collection (ATCC; 10801 University Boulevard, Manassas, Va. 20110-2209) and can be chosen to ensure the correct modification and processing of the foreign protein. [0158]
  • Stable expression is preferred for long-term, high-yield production of recombinant proteins. For example, cell lines which stably express protein kinase-like polypeptides can be transformed using expression vectors which can contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells can be allowed to grow for 1-2 days in an enriched medium before they are switched to a selective medium. The purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells which successfully express the introduced protein kinase-like protein sequences. Resistant clones of stably transformed cells can be proliferated using tissue culture techniques appropriate to the cell type. See, for example, A[0159] NIMAL CELL CULTURE, R. I. Freshney, ed., 1986.
  • Any number of selection systems can be used to recover transformed cell lines. [0160]
  • These include, but are not limited to, the herpes simplex virus thymidine kinase (Wigler et al., [0161] Cell 11, 223-32, 1977) and adenine phosphoribosyltransferase (Lowy et al., Cell 22, 817-23, 1980) genes which can be employed in tk or aprt(cells, respectively. Also, antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection. For example, dhfr confers resistance to methotrexate (Wigler et al., Proc. Natl. Acad. Sci. 77, 3567-70, 1980), npt confers resistance to the aminoglycosides, neomycin and G-418 (Colbere-Garapin et al., J. Mol. Biol. 150, 1-14, 1981), and als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (Murray, 1992, supra). Additional selectable genes have been described. For example, trpB allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine (Hartman & Mulligan, Proc. Natl. Acad. Sci. 85, 8047-51, 1988). Visible markers such as anthocyanins, β-glucuronidase and its substrate GUS, and luciferase and its substrate luciferin, can be used to identify transformants and to quantify the amount of transient or stable protein expression attributable to a specific vector system (Rhodes et al., Methods Mol. Biol. 55, 121-131, 1995).
  • Detecting Expression [0162]
  • Although the presence of marker gene expression suggests that the protein kinase-like polynucleotide is also present, its presence and expression may need to be confirmed. For example, if a sequence encoding a protein kinase-like polypeptide is inserted within a marker gene sequence, transformed cells containing sequences that encode a protein kinase-like polypeptide can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding a protein kinase-like polypeptide under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the protein kinase-like polynucleotide. [0163]
  • Alternatively, host cells which contain a protein kinase-like polynucleotide and which express a protein kinase-like polypeptide can be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations and protein bioassay or immunoassay techniques that include membrane, solution, or chip-based technologies for the detection and/or quantification of nucleic acid or protein. For example, the presence of a polynucleotide sequence encoding a protein kinase-like polypeptide can be detected by DNA-DNA or DNA-RNA hybridization or amplification using probes or fragments or fragments of polynucleotides encoding a protein kinase-like polypeptide. Nucleic acid amplification-based assays involve the use of oligonucleotides selected from sequences encoding a protein kinase-like polypeptide to detect transformants that contain a protein kinase-like polynucleotide. [0164]
  • A variety of protocols for detecting and measuring the expression of a protein kinase-like polypeptide, using either polyclonal or monoclonal antibodies specific for the polypeptide, are known in the art. Examples include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence activated cell sorting (FACS). A two-site, monoclonal-based immunoassay using monoclonal antibodies reactive to two non-interfering epitopes on a protein kinase-like polypeptide can be used, or a competitive binding assay can be employed. These and other assays are described in Hampton et al., S[0165] EROLOGICAL METHODS: A LABORATORY MANUAL, APS Press, St. Paul, Minn., 1990) and Maddox et al., J. Exp. Med. 158, 1211-1216, 1983).
  • A wide variety of labels and conjugation techniques are known by those skilled in the art and can be used in various nucleic acid and amino acid assays. Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding protein kinase-like polypeptides include oligolabeling, nick translation, end-labeling, or PCR amplification using a labeled nucleotide. Alternatively, sequences encoding a protein kinase-like polypeptide can be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and can be used to synthesize RNA probes in vitro by addition of labeled nucleotides and an appropriate RNA polymerase such as T7, T3, or SP6. These procedures can be conducted using a variety of commercially available kits (Amersham Pharmacia Biotech, Promega, and US Biochemical). Suitable reporter molecules or labels which can be used for ease of detection include radionuclides, enzymes, and fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like. [0166]
  • Expression and Purification of Polypeptides [0167]
  • Host cells transformed with nucleotide sequences encoding a protein kinase-like polypeptide can be cultured under conditions suitable for the expression and recovery of the protein from cell culture. The polypeptide produced by a transformed cell can be secreted or contained intracellularly depending on the sequence and/or the vector used. As will be understood by those of skill in the art, expression vectors containing polynucleotides which encode protein kinase-like polypeptides can be designed to contain signal sequences which direct secretion of soluble protein kinase-like polypeptides through a prokaryotic or eukaryotic cell membrane or which direct the membrane insertion of membrane-bound protein kinase-like polypeptide. [0168]
  • As discussed above, other constructions can be used to join a sequence encoding a protein kinase-like polypeptide to a nucleotide sequence encoding a polypeptide domain which will facilitate purification of soluble proteins. Such purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, Wash.). Inclusion of cleavable linker sequences such as those specific for Factor Xa or enterokinase (Invitrogen, San Diego, Calif.) between the purification domain and the protein kinase-like polypeptide also can be used to facilitate purification. One such expression vector provides for expression of a fusion protein containing a protein kinase-like polypeptide and 6 histidine residues preceding a thioredoxin or an enterokinase cleavage site. The histidine residues facilitate purification by IMAC (immobilized metal ion affinity chromatography, as described in Porath et al., [0169] Prot. Exp. Purif. 3, 263-281, 1992), while the enterokinase cleavage site provides a means for purifying the protein kinase-like polypeptide from the fusion protein. Vectors that contain fusion proteins are disclosed in Kroll et al., DNA Cell Biol. 12,441453,1993.
  • Chemical Synthesis [0170]
  • Sequences encoding a protein kinase-like polypeptide can be synthesized, in whole or in part, using chemical methods well known in the art (see Caruthers et al., [0171] Nucl. Acids Res. Symp. Ser. 215-223, 1980; Horn et al. Nucl. Acids Res. Symp. Ser. 225-232, 1980). Alternatively, a protein kinase-like polypeptide itself can be produced using chemical methods to synthesize its amino acid sequence, such as by direct peptide synthesis using solid-phase techniques (Merrifield, J. Am. Chem. Soc. 85, 2149-2154, 1963; Roberge et al., Science 269, 202-204, 1995). Protein synthesis can be performed using manual techniques or by automation. Automated synthesis can be achieved, for example, using Applied Biosystems 431A Peptide Synthesizer (Perkin Elmer). Optionally, fragments of protein kinase-like polypeptides can be separately synthesized and combined using chemical methods to produce a full-length molecule.
  • The newly synthesized peptide can be substantially purified by preparative high performance liquid chromatography (e.g., Creighton, P[0172] ROTEINS: STRUCTURES AND MOLECULAR PRINCIPLES, WH Freeman and Co., New York, N.Y., 1983). The composition of a synthetic protein kinase-like polypeptide can be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure; see Creighton, supra). Additionally, any portion of the amino acid sequence of the protein kinase-like polypeptide can be altered during direct synthesis and/or combined using chemical methods with sequences from other proteins to produce a variant polypeptide or a fusion protein.
  • Production of Altered Polypeptides [0173]
  • As will be understood by those of skill in the art, it may be advantageous to produce protein kinase-like polypeptide-encoding nucleotide sequences possessing non-naturally occurring codons. For example, codons preferred by a particular prokaryotic or eukaryotic host can be selected to increase the rate of protein expression or to produce an RNA transcript having desirable properties, such as a half-life that is longer than that of a transcript generated from the naturally occurring sequence. [0174]
  • The nucleotide sequences disclosed herein can be engineered using methods generally known in the art to alter protein kinase-like polypeptide-encoding sequences for a variety of reasons, including but not limited to, alterations which modify the cloning, processing, and/or expression of the polypeptide or mRNA product. DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides can be used to engineer the nucleotide sequences. For example, site-directed mutagenesis can be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, introduce mutations, and so forth. [0175]
  • Antibodies [0176]
  • Any type of antibody known in the art can be generated to bind specifically to an epitope of a protein kinase-like polypeptide. “Antibody” as used herein includes intact immunoglobulin molecules, as well as fragments thereof, such as Fab, F(ab′)[0177] 2, and Fv, which are capable of binding an epitope of a protein kinase-like polypeptide.
  • Typically, at least 6, 8, 10, or 12 contiguous amino acids are required to form an epitope. However, epitopes which involve non-contiguous amino acids may require more, e.g., at least 15, 25, or 50 amino acids. [0178]
  • An antibody which specifically binds to an epitope of a protein kinase-like polypeptide can be used therapeutically, as well as in immunochemical assays, such as Western blots, ELISAs, radioimmunoassays, immunohistochemical assays, immunoprecipitations, or other immunochemical assays known in the art. Various immunoassays can be used to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays are well known in the art. Such immunoassays typically involve the measurement of complex formation between an immunogen and an antibody that specifically binds to the immunogen. [0179]
  • Typically, an antibody which specifically binds to a protein kinase-like polypeptide provides a detection signal at least 5-, 10-, or 20-fold higher than a detection signal provided with other proteins when used in an immunochemical assay. Preferably, antibodies which specifically bind to protein kinase-like polypeptides do not detect other proteins in immunochemical assays and can immunoprecipitate a protein kinase-like polypeptide from solution. [0180]
  • Human protein kinase-like polypeptides can be used to immunize a mammal, such as a mouse, rat, rabbit, guinea pig, monkey, or human, to produce polyclonal antibodies. If desired, a protein kinase-like polypeptide can be conjugated to a carrier protein, such as bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin. Depending on the host species, various adjuvants can be used to increase the immunological response. Such adjuvants include, but are not limited to, Freund's adjuvant, mineral gels (e.g., aluminum hydroxide), and surface active substances (e.g. lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol). Among adjuvants used in humans, BCG (bacilli Calmette-Guerin) and [0181] Corynebacterium parvum are especially useful.
  • Monoclonal antibodies that specifically bind to a protein kinase-like polypeptide can be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These techniques include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique (Kohler et al., [0182] Nature 256, 495-497, 1985; Kozbor et al., J. Immunol. Methods 81, 31-42, 1985; Cote et al., Proc. Natl. Acad. Sci. 80, 2026-2030, 1983; Cole et al., Mol. Cell Biol. 62, 109-120, 1984).
  • In addition, techniques developed for the production of “chimeric antibodies,” the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used (Morrison et al., [0183] Proc Natl. Acad. Sci. 81, 6851-6855, 1984; Neuberger et al., Nature 312, 604-608, 1984; Takeda et al., Nature 314, 452-454, 1985). Monoclonal and other antibodies also can be “humanized” to prevent a patient from mounting an immune response against the antibody when it is used therapeutically. Such antibodies may be sufficiently similar in sequence to human antibodies to be used directly in therapy or may require alteration of a few key residues. Sequence differences between rodent antibodies and human sequences can be minimized by replacing residues which differ from those in the human sequences by site directed mutagenesis of individual residues or by grating of entire complementarity determining regions. Alternatively, humanized antibodies can be produced using recombinant methods, as described in GB2188638B. Antibodies that specifically bind to a protein kinase-like polypeptide can contain antigen binding sites which are either partially or fully humanized, as disclosed in U.S. Pat. No. 5,565,332.
  • Alternatively, techniques described for the production of single chain antibodies can be adapted using methods known in the art to produce single chain antibodies that specifically bind to protein kinase-like polypeptides. Antibodies with related specificity, but of distinct idiotypic composition, can be generated by chain shuffling from random combinatorial immunoglobin libraries (Burton, [0184] Proc. Natl. Acad. Sci. 88, 11120-23, 1991).
  • Single-chain antibodies also can be constructed using a DNA amplification method, such as PCR, using hybridoma cDNA as a template (Thirion et al., 1996[0185] , Eur. J. Cancer Prev. 5, 507-11). Single-chain antibodies can be mono- or bispecific, and can be bivalent or tetravalent. Construction of tetravalent, bispecific single-chain antibodies is taught, for example, in Coloma & Morrison, 1997, Nat. Biotechnol. 15, 159-63. Construction of bivalent, bispecific single-chain antibodies is taught in Mallender & Voss, 1994, J. Biol. Chem. 269, 199-206.
  • A nucleotide sequence encoding a single-chain antibody can be constructed using manual or automated nucleotide synthesis, cloned into an expression construct using standard recombinant DNA methods, and introduced into a cell to express the coding sequence, as described below. Alternatively, single-chain antibodies can be produced directly using, for example, filamentous phage technology (Verhaar et al., 1995[0186] , Int. J. Cancer 61, 497-501; Nicholls et al., 1993, J. Immunol. Meth. 165, 81-91).
  • Antibodies which specifically bind to protein kinase-like polypeptides also can be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature (Orlandi et al., [0187] Proc. Natl. Acad. Sci. 86, 3833-3837, 1989; Winter et al., Nature 349, 293-299, 1991).
  • Other types of antibodies can be constructed and used therapeutically in methods of the invention. For example, chimeric antibodies can be constructed as disclosed in WO 93/03151. Binding proteins which are derived from immunoglobulins and which are multivalent and multispecific, such as the “diabodies” described in WO 94/13804, also can be prepared. [0188]
  • Antibodies according to the invention can be purified by methods well known in the art. For example, antibodies can be affinity purified by passage over a column to which a protein kinase-like polypeptide is bound. The bound antibodies can then be eluted from the column using a buffer with a high salt concentration. [0189]
  • Antisense Oligonucleotides [0190]
  • Antisense oligonucleotides are nucleotide sequences that are complementary to a specific DNA or RNA sequence. Once introduced into a cell, the complementary nucleotides combine with natural sequences produced by the cell to form complexes and block either transcription or translation. Preferably, an antisense oligonucleotide is at least 11 nucleotides in length, but can be at least 12, 15, 20, 25, 30, 35, 40, 45, or 50 or more nucleotides long. Longer sequences also can be used. Antisense oligonucleotide molecules can be provided in a DNA construct and introduced into a cell as described above to decrease the level of protein kinase-like gene products in the cell. [0191]
  • Antisense oligonucleotides can be deoxyribonucleotides, ribonucleotides, or a combination of both. Oligonucleotides can be synthesized manually or by an automated synthesizer, by covalently linking the 5′ end of one nucleotide with the 3′ end of another nucleotide with non-phosphodiester internucleotide linkages such alkyl-phosphonates, phosphorothioates, phosphorodithioates, alkylphosphonothioates, alkylphosphonates, phosphoramidates, phosphate esters, carbamates, acetamidate, carboxymethyl esters, carbonates, and phosphate triesters. See Brown, [0192] Meth. Mol. Biol. 20, 1-8, 1994; Sonveaux, Meth. Mol. Biol. 26, 1-72, 1994; Uhlmann et al., Chem. Rev. 90, 543-583,1990.
  • Modifications of protein kinase-like gene expression can be obtained by designing antisense oligonucleotides that will form duplexes to the control, 5′, or regulatory regions of the protein kinase-like gene. Oligonucleotides derived from the transcription initiation site, e.g., between positions −10 and +10 from the start site, are preferred. Similarly, inhibition can be achieved using “triple helix” base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or chaperons. Therapeutic advances using triplex DNA have been described in the literature (e.g., Gee et al., in Huber & Carr, M[0193] OLECULAR AND IMMUNOLOGIC APPROACHES, Futura Publishing Co., Mt. Kisco, N.Y., 1994). An antisense oligonucleotide also can be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.
  • Precise complementarity is not required for successful complex formation between an antisense oligonucleotide and the complementary sequence of a protein kinase-like polynucleotide. Antisense oligonucleotides which comprise, for example, 2, 3, 4, or 5 or more stretches of contiguous nucleotides which are precisely complementary to a protein kinase-like polynucleotide, each separated by a stretch of contiguous nucleotides which are not complementary to adjacent protein kinase-like protein nucleotides, can provide sufficient targeting specificity for protein kinase-like mRNA. Preferably, each stretch of complementary contiguous nucleotides is at least 4, 5, 6, 7, or 8 or more nucleotides in length. Non-complementary intervening sequences are preferably 1, 2, 3, or 4 nucleotides in length. One skilled in the art can easily use the calculated melting point of an antisense-sense pair to determine the degree of mismatching which will be tolerated between a particular antisense oligonucleotide and a particular protein kinase-like polynucleotide sequence. [0194]
  • Antisense oligonucleotides can be modified without affecting their ability to hybridize to a protein kinase-like polynucleotide. These modifications can be internal or at one or both ends of the antisense molecule. For example, internucleoside phosphate linkages can be modified by adding cholesteryl or diamine moieties with varying numbers of carbon residues between the amino groups and terminal ribose. Modified bases and/or sugars, such as arabinose instead of ribose, or a 3′,5′-substituted oligonucleotide in which the 3′ hydroxyl group or the 5′ phosphate group are substituted, also can be employed in a modified antisense oligonucleotide. These modified oligonucleotides can be prepared by methods well known in the art. See, e.g., Agrawal et al., [0195] Trends Biotechnol. 10, 152-158, 1992; Uhlmann et al., Chem. Rev. 90, 543-584, 1990; Uhlmann et al., Tetrahedron. Lett. 215, 3539-3542, 1987.
  • Ribozymes [0196]
  • Ribozymes are RNA molecules with catalytic activity. See, e.g., Cech, [0197] Science 236, 1532-1539; 1987; Cech, Ann. Rev. Biochem. 59, 543-568; 1990, Cech, Curr. Opin. Struct. Biol. 2, 605-609; 1992, Couture & Stinchcomb, Trends Genet. 12, 510-515, 1996. Ribozymes can be used to inhibit gene function by cleaving an RNA sequence, as is known in the art (e.g., Haseloff et al., U.S. Pat. No. 5,641,673). The mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. Examples include engineered hammerhead motif ribozyme molecules that can specifically and efficiently catalyze endonucleolytic cleavage of specific nucleotide sequences.
  • The coding sequence of a protein kinase-like polynucleotide can be used to generate ribozymes that will specifically bind to mRNA transcribed from the protein kinase-like polynucleotide. Methods of designing and constructing ribozymes which can cleave other RNA molecules in trans in a highly sequence specific manner have been developed and described in the art (see Haseloff et al. [0198] Nature 334, 585-591, 1988). For example, the cleavage activity of ribozymes can be targeted to specific RNAs by engineering a discrete “hybridization” region into the ribozyme. The hybridization region contains a sequence complementary to the target RNA and thus specifically hybridizes with the target (see, for example, Gerlach et al., EP 321,201).
  • Specific ribozyme cleavage sites within a protein kinase-like protein RNA target can be identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences: GUA, GUU, and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides corresponding to the region of the target RNA containing the cleavage site can be evaluated for secondary structural features which may render the target inoperable. Suitability of candidate protein kinase-like protein RNA targets also can be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays. Longer complementary sequences can be used to increase the affinity of the hybridization sequence for the target. The hybridizing and cleavage regions of the ribozyme can be integrally related such that upon hybridizing to the target RNA through the complementary regions, the catalytic region of the ribozyme can cleave the target. [0199]
  • Ribozymes can be introduced into cells as part of a DNA construct. Mechanical methods, such as microinjection, liposome-mediated transfection, electroporation, or calcium phosphate precipitation, can be used to introduce a ribozyme-containing DNA construct into cells in which it is desired to decrease protein kinase-like protein expression. Alternatively, if it is desired that the cells stably retain the DNA construct, the construct can be supplied on a plasmid and maintained as a separate element or integrated into the genome of the cells, as is known in the art. A ribozyme-encoding DNA construct can include transcriptional regulatory elements, such as a promoter element, an enhancer or UAS element, and a transcriptional terminator signal, for controlling transcription of ribozymes in the cells. [0200]
  • As taught in Haseloff et al., U.S. Pat. No. 5,641,673, ribozymes can be engineered so that ribozyme expression will occur in response to factors that induce expression of a target gene. Ribozymes also can be engineered to provide an additional level of regulation, so that destruction of mRNA occurs only when both a ribozyme and a target gene are induced in the cells. [0201]
  • Differentially Expressed Genes [0202]
  • Described herein are methods for the identification of genes whose products interact with human protein kinase-like protein. Such genes may represent genes that are differentially expressed in disorders including, but not limited to, cancer, CNS disorders, COPD, obesity, diabetes, and cardiovascular disorders [0203]
  • Further, such genes may represent genes that are differentially regulated in response to manipulations relevant to the progression or treatment of such diseases. Additionally, such genes may have a temporally modulated expression, increased or decreased at different stages of tissue or organism development. A differentially expressed gene may also have its expression modulated under control versus experimental conditions. In addition, the human protein kinase-like gene or gene product may itself be tested for differential expression. [0204]
  • The degree to which expression differs in a normal versus a diseased state need only be large enough to be visualized via standard characterization techniques such as differential display techniques. Other such standard characterization techniques by which expression differences may be visualized include but are not limited to, quantitative RT (reverse transcriptase), PCR, and Northern analysis. [0205]
  • Identification of Differentially Expressed Genes [0206]
  • To identify differentially expressed genes total RNA or, preferably, mRNA is isolated from tissues of interest. For example, RNA samples are obtained from tissues of experimental subjects and from corresponding tissues of control subjects. Any RNA isolation technique that does not select against the isolation of mRNA may be utilized for the purification of such RNA samples. See, for example, Ausubel et al., ed., C[0207] URRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, Inc. New York, 1987-1993. Large numbers of tissue samples may readily be processed using techniques well known to those of skill in the art, such as, for example, the single-step RNA isolation process of Chomczynski, U.S. Pat. No. 4,843,155.
  • Transcripts within the collected RNA samples that represent RNA produced by differentially expressed genes are identified by methods well known to those of skill in the art. They include, for example, differential screening (Tedder et al., [0208] Proc. Natl. Acad. Sci. U.S.A. 85, 208-12, 1988), subtractive hybridization (Hedrick et al., Nature 308, 149-53; Lee et al., Proc. Natl. Acad. Sci. U.S.A. 88, 2825, 1984), and, preferably, differential display (Liang & Pardee, Science 257, 967-71, 1992; U.S. Pat. No. 5,262,311).
  • The differential expression information may itself suggest relevant methods for the treatment of disorders involving the human protein kinase-like protein. For example, treatment may include a modulation of expression of the differentially expressed genes and/or the gene encoding the human protein kinase-like protein. The differential expression information may indicate whether the expression or activity of the differentially expressed gene or gene product or the human protein kinase-like gene or gene product are up-regulated or down-regulated. [0209]
  • Screening Methods [0210]
  • The invention provides assays for screening test compounds that bind to or modulate the activity of a protein kinase-like polypeptide or a protein kinase-like polynucleotide. A test compound preferably binds to a protein kinase-like polypeptide or polynucleotide. More preferably, a test compound decreases or increases enzyme activity by at least about 10, preferably about 50, more preferably about 75, 90, or 100% relative to the absence of the test compound. [0211]
  • Test Compounds [0212]
  • Test compounds can be pharmacologic agents already known in the art or can be compounds previously unknown to have any pharmacological activity. The compounds can be naturally occurring or designed in the laboratory. They can be isolated from microorganisms, animals, or plants, and can be produced recombinantly, or synthesized by chemical methods known in the art. If desired, test compounds can be obtained using any of the numerous combinatorial library methods known in the art, including but not limited to, biological libraries, spatially addressable parallel solid phase or solution phase libraries, synthetic library methods requiring deconvolution, the “one-bead one-compound” library method, and synthetic library methods using affinity chromatography selection. The biological library approach is limited to polypeptide libraries, while the other four approaches are applicable to polypeptide, non-peptide oligomer, or small molecule libraries of compounds. See Lam, [0213] Anticancer Drug Des. 12, 145, 1997.
  • Methods for the synthesis of molecular libraries are well known in the art (see, for example, DeWitt et al., [0214] Proc. Natl. Acad. Sci. U.S.A. 90, 6909, 1993; Erb et al. Proc. Natl. Acad. Sci. U.S.A. 91, 11422, 1994; Zuckermann et al., J. Med. Chem. 37, 2678, 1994; Cho et al., Science 261, 1303, 1993; Carell et al., Angew. Chem. Int. Ed. Engl. 33, 2059, 1994; Carell et al., Angew. Chem. Int. Ed. Engl. 33, 2061; Gallop et al., J. Med. Chem. 37, 1233, 1994). Libraries of compounds can be presented in solution (see, e.g., Houghten, BioTechniques 13, 412-421, 1992), or on beads (Lam, Nature 354, 82-84, 1991), chips (Fodor, Nature 364, 555-556, 1993), bacteria or spores (Ladner, U.S. Pat. No. 5,223,409), plasmids (Cull et al., Proc. Natl. Acad. Sci. U.S.A. 89, 1865-1869, 1992), or phage (Scott & Smith, Science 249, 386-390, 1990; Devlin, Science 249, 404-406, 1990); Cwirla et al., Proc. Natl. Acad. Sci. 97, 6378-6382, 1990; Felici, J. Mol. Biol. 222, 301-310, 1991; and Ladner, U.S. Pat. No. 5,223,409).
  • High Throughput Screening [0215]
  • Test compounds can be screened for the ability to bind to protein kinase-like polypeptides or polynucleotides or to affect protein kinase-like protein activity or protein kinase-like gene expression using high throughput screening. Using high throughput screening, many discrete compounds can be tested in parallel so that large numbers of test compounds can be quickly screened. The most widely established techniques utilize 96-well microfiter plates. The wells of the microtiter plates typically require assay volumes that range from 50 to 500 μl. In addition to the plates, many instruments, materials, pipettors, robotics, plate washers, and plate readers are commercially available to fit the 96-well format. [0216]
  • Alternatively, “free format assays,” or assays that have no physical barrier between samples, can be used. For example, an assay using pigment cells (melanocytes) in a simple homogeneous assay for combinatorial peptide libraries is described by Jayawickreme et al., [0217] Proc. Natl. Acad. Sci. U.S.A. 19, 1614-18 (1994). The cells are placed under agarose in petri dishes, then beads that carry combinatorial compounds are placed on the surface of the agarose. The combinatorial compounds are partially released the compounds from the beads. Active compounds can be visualized as dark pigment areas because, as the compounds diffuse locally into the gel matrix, the active compounds cause the cells to change colors.
  • Another example of a free format assay is described by Chelsky, “Strategies for Screening Combinatorial Libraries: Novel and Traditional Approaches,” reported at the First Annual Conference of The Society for Biomolecular Screening in Philadelphia, Pa. (Nov. 7-10, 1995). Chelsky placed a simple homogenous enzyme assay for carbonic anhydrase inside an agarose gel such that the enzyme in the gel would cause a color change throughout the gel. Thereafter, beads carrying combinatorial compounds via a photolinker were placed inside the gel and the compounds were partially released by UV-light. Compounds that inhibited the enzyme were observed as local zones of inhibition having less color change. [0218]
  • Yet another example is described by Salmon et al., [0219] Molecular Diversity 2, 57-63 (1996). In this example, combinatorial libraries were screened for compounds that had cytotoxic effects on cancer cells growing in agar.
  • Another high throughput screening method is described in Beutel et al., U.S. Pat. No. 5,976,813. In this method, test samples are placed in a porous matrix. One or more assay components are then placed within, on top of, or at the bottom of a matrix such as a gel, a plastic sheet, a filter, or other form of easily manipulated solid support. When samples are introduced to the porous matrix they diffuse sufficiently slowly, such that the assays can be performed without the test samples running together. [0220]
  • Binding Assays [0221]
  • For binding assays, the test compound is preferably a small molecule that binds to and occupies, for example, the active site of the protein kinase-like polypeptide, such that normal biological activity is prevented. Examples of such small molecules include, but are not limited to, small peptides or peptide-like molecules. [0222]
  • In binding assays, either the test compound or the protein kinase-like polypeptide can comprise a detectable label, such as a fluorescent, radioisotopic, chemiluminescent, or enzymatic label, such as horseradish peroxidase, alkaline phosphatase, or luciferase. Detection of a test compound that is bound to the protein kinase-like polypeptide can then be accomplished, for example, by direct counting of radio-emmission, by scintillation counting, or by determining conversion of an appropriate substrate to a detectable product. [0223]
  • Alternatively, binding of a test compound to a protein kinase-like polypeptide can be determined without labeling either of the interactants. For example, a microphysiometer can be used to detect binding of a test compound with a protein kinase-like polypeptide. A microphysiometer (e.g., Cytosensor™) is an analytical instrument that measures the rate at which a cell acidifies its environment using a light-addressable potentiometric sensor (LAPS). Changes in this acidification rate can be used as an indicator of the interaction between a test compound and a protein kinase-like polypeptide (McConnell et al., [0224] Science 257, 1906-1912, 1992).
  • Determining the ability of a test compound to bind to a protein kinase-like polypeptide also can be accomplished using a technology such as real-time Bimolecular Interaction Analysis (BIA) (Sjolander & Urbaniczky, [0225] Anal. Chem. 63, 2338-2345, 1991, and Szabo et al., Curr. Opin. Struct. Biol. 5, 699-705, 1995). BIA is a technology for studying biospecific interactions in real time, without labeling any of the interactants (e.g., BIAcore™). Changes in the optical phenomenon surface plasmon resonance (SPR) can be used as an indication of real-time reactions between biological molecules.
  • In yet another aspect of the invention, a protein kinase-like polypeptide can be used as a “bait protein” in a two-hybrid assay or three-hybrid assay (see, e.g., U.S. Pat. No. 5,283,317; Zervos et al., [0226] Cell 72, 223-232, 1993; Madura et al., J. Biol. Chem. 268, 12046-12054, 1993; Bartel et al., BioTechniques 14, 920-924, 1993; Iwabuchi et al., Oncogene 8, 1693-1696, 1993; and Brent WO94/10300), to identify other proteins which bind to or interact with the protein kinase-like polypeptide and modulate its activity.
  • The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs. For example, in one construct, polynucleotide encoding a protein kinase-like polypeptide can be fused to a polynucleotide encoding the DNA binding domain of a known transcription factor (e.g., GAL-4). In the other construct a DNA sequence that encodes an unidentified protein (“prey” or “sample”) can be fused to a polynucleotide that codes for the activation domain of the known transcription factor. If the “bait” and the “prey” proteins are able to interact in vivo to form an protein-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ), which is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected, and cell colonies containing the functional transcription factor can be isolated and used to obtain the DNA sequence encoding the protein that interacts with the protein kinase-like polypeptide. [0227]
  • It may be desirable to immobilize either the protein kinase-like polypeptide (or polynucleotide) or the test compound to facilitate separation of bound from unbound forms of one or both of the interactants, as well as to accommodate automation of the assay. Thus, either the protein kinase-like polypeptide (or polynucleotide) or the test compound can be bound to a solid support. Suitable solid supports include, but are not limited to, glass or plastic slides, tissue culture plates, microtiter wells, tubes, silicon chips, or particles such as beads (including, but not limited to, latex, polystyrene, or glass beads). Any method known in the art can be used to attach the enzyme polypeptide (or polynucleotide) or test compound to a solid support, including use of covalent and non-covalent linkages, passive absorption, or pairs of binding moieties attached respectively to the polypeptide (or polynucleotide) or test compound and the solid support. Test compounds are preferably bound to the solid support in an array, so that the location of individual test compounds can be tracked. Binding of a test compound to a protein kinase-like polypeptide (or polynucleotide) can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and microcentrifuge tubes. [0228]
  • In one embodiment, the protein kinase-like polypeptide is a fusion protein comprising a domain that allows the protein kinase-like polypeptide to be bound to a solid support. For example, glutathione-S-transferase fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, Mo.) or glutathione derivatized microtiter plates, which are then combined with the test compound or the test compound and the non-adsorbed protein kinase-like polypeptide; the mixture is then incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components. Binding of the interactants can be determined either directly or indirectly, as described above. Alternatively, the complexes can be dissociated from the solid support before binding is determined. [0229]
  • Other techniques for immobilizing proteins or polynucleotides on a solid support also can be used in the screening assays of the invention. For example, either a protein kinase-like polypeptide (or polynucleotide) or a test compound can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated protein kinase-like polypeptides (or polynucleotides) or test compounds can be prepared from biotin-NHS(N-hydroxysuccinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, Ill.) and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies which specifically bind to a protein kinase-like polypeptide, polynucleotide, or a test compound, but which do not interfere with a desired binding site, such as the active site of the protein kinase-like polypeptide, can be derivatized to the wells of the plate. Unbound target or protein can be trapped in the wells by antibody conjugation. [0230]
  • Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies which specifically bind to the protein kinase-like polypeptide or test compound, enzyme-linked assays which rely on detecting an activity of the protein kinase-like polypeptide, and SDS gel electrophoresis under non-reducing conditions. [0231]
  • Screening for test compounds which bind to a protein kinase-like polypeptide or polynucleotide also can be carried out in an intact cell. Any cell which comprises a protein kinase-like polypeptide or polynucleotide can be used in a cell-based assay system. A protein kinase-like polynucleotide can be naturally occurring in the cell or can be introduced using techniques such as those described above. Binding of the test compound to a protein kinase-like polypeptide or polynucleotide is determined as described above. [0232]
  • Enzyme Assays [0233]
  • Test compounds can be tested for the ability to increase or decrease the protein kinase activity of a human protein kinase-like polypeptide. Protein kinase activity can be measured, for example, as described in U.S. Patent U.S. Pat. No. 6,194,186 [0234]
  • Enzyme assays can be carried out after contacting either a purified protein kinase-like polypeptide, a cell membrane preparation, or an intact cell with a test compound. A test compound that decreases a protein kinase activity of a protein kinase-like polypeptide by at least about 10, preferably about 50, more preferably about 75, 90, or 100% is identified as a potential therapeutic agent for decreasing protein kinase-like protein activity. A test compound which increases a protein kinase activity of a human protein kinase-like polypeptide by at least about 10, preferably about 50, more preferably about 75, 90, or 100% is identified as a potential therapeutic agent for increasing human protein kinase-like protein activity. [0235]
  • Gene Expression [0236]
  • In another embodiment, test compounds that increase or decrease protein kinase-like gene expression are identified. A protein kinase-like polynucleotide is contacted with a test compound, and the expression of an RNA or polypeptide product of the protein kinase-like polynucleotide is determined. The level of expression of appropriate mRNA or polypeptide in the presence of the test compound is compared to the level of expression of mRNA or polypeptide in the absence of the test compound. The test compound can then be identified as a modulator of expression based on this comparison. For example, when expression of mRNA or polypeptide is greater in the presence of the test compound than in its absence, the test compound is identified as a stimulator or enhancer of the mRNA or polypeptide expression. Alternatively, when expression of the mRNA or polypeptide is less in the presence of the test compound than in its absence, the test compound is identified as an inhibitor of the mRNA or polypeptide expression. [0237]
  • The level of protein kinase-like mRNA or polypeptide expression in the cells can be determined by methods well known in the art for detecting mRNA or polypeptide. Either qualitative or quantitative methods can be used. The presence of polypeptide products of a protein kinase-like polynucleotide can be determined, for example, using a variety of techniques known in the art, including immunochemical methods such as radioimmunoassay, Western blotting, and immunohistochemistry. Alternatively, polypeptide synthesis can be determined in vivo, in a cell culture, or in an in vitro translation system by detecting incorporation of labeled amino acids into a protein kinase-like polypeptide. [0238]
  • Such screening can be carried out either in a cell-free assay system or in an intact cell. Any cell that expresses a protein kinase-like polynucleotide can be used in a cell-based assay system. The protein kinase-like polynucleotide can be naturally occurring in the cell or can be introduced using techniques such as those described above. Either a primary culture or an established cell line, such as CHO or human embryonic kidney 293 cells, can be used. [0239]
  • Pharmaceutical Compositions [0240]
  • The invention also provides pharmaceutical compositions that can be administered to a patient to achieve a therapeutic effect. Pharmaceutical compositions of the invention can comprise, for example, a protein kinase-like polypeptide, protein kinase-like polynucleotide, ribozymes or antisense oligonucleotides, antibodies which specifically bind to a protein kinase-like polypeptide, or mimetics, activators, or inhibitors of a protein kinase-like polypeptide activity. The compositions can be administered alone or in combination with at least one other agent, such as stabilizing compound, which can be administered in any sterile, biocompatible pharmaceutical carrier, including, but not limited to, saline, buffered saline, dextrose, and water. The compositions can be administered to a patient alone, or in combination with other agents, drugs or hormones. [0241]
  • In addition to the active ingredients, these pharmaceutical compositions can contain suitable pharmaceutically-acceptable carriers comprising excipients and auxiliaries that facilitate processing of the active compounds into preparations which can be used pharmaceutically. Pharmaceutical compositions of the invention can be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intraperitoneal, intranasal, parenteral, topical, sublingual, or rectal means. Pharmaceutical compositions for oral administration can be formulated using pharmaceutically acceptable carriers well known in the art in dosages suitable for oral administration. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for ingestion by the patient. [0242]
  • Pharmaceutical preparations for oral use can be obtained through combination of active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are carbohydrate or protein fillers, such as sugars, including lactose, sucrose, mannitol, or sorbitol; starch from corn, wheat, rice, potato, or other plants; cellulose, such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethylcellulose; gums including arabic and tragacanth; and proteins such as gelatin and collagen. If desired, disintegrating or solubilizing agents can be added, such as the cross-linked polyvinyl pyrrolidone, agar, alginic acid, or a salt thereof, such as sodium alginate. [0243]
  • Dragee cores can be used in conjunction with suitable coatings, such as concentrated sugar solutions, which also can contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments can be added to the tablets or dragee coatings for product identification or to characterize the quantity of active compound, i.e., dosage. [0244]
  • Pharmaceutical preparations that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating, such as glycerol or sorbitol. Push-fit capsules can contain active ingredients mixed with a filler or binders, such as lactose or starches, lubricants, such as talc or magnesium stearate, and, optionally, stabilizers. In soft capsules, the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid, or liquid polyethylene glycol with or without stabilizers. [0245]
  • Pharmaceutical formulations suitable for parenteral administration can be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiologically buffered saline. Aqueous injection suspensions can contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Additionally, suspensions of the active compounds can be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Non-lipid polycationic amino polymers also can be used for delivery. Optionally, the suspension also can contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. For topical or nasal administration, penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. [0246]
  • The pharmaceutical compositions of the present invention can be manufactured in a manner that is known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes. The pharmaceutical composition can be provided as a salt and can be formed with many acids, including but not limited to, hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms. In other cases, the preferred preparation can be a lyophilized powder which can contain any or all of the following: 1-50 mM histidine, 0.1%-2% sucrose, and 2-7% mannitol, at a pH range of 4.5 to 5.5, that is combined with buffer prior to use. [0247]
  • Further details on techniques for formulation and administration can be found in the latest edition of REMINGTON'S PHARMACEUTICAL SCENCES (Maack Publishing Co., Easton, Pa.). After pharmaceutical compositions have been prepared, they can be placed in an appropriate container and labeled for treatment of an indicated condition. Such labeling would include amount, frequency, and method of administration. [0248]
  • Therapeutic Indications and Methods [0249]
  • Human protein kinase-like protein can be regulated to treat cancer, CNS disorders, COPD, obesity, diabetes, and cardiovascular disorders. [0250]
  • Cancer [0251]
  • Cancer is a disease fundamentally caused by oncogenic cellular transformation. There are several hallmarks of transformed cells that distinguish them from their normal counterparts and underlie the pathophysiology of cancer. These include uncontrolled cellular proliferation, unresponsiveness to normal death-inducing signals (immortalization), increased cellular motility and invasiveness, increased ability to recruit blood supply through induction of new blood vessel formation (angiogenesis), genetic instability, and dysregulated gene expression. Various combinations of these aberrant physiologies, along with the acquisition of drug-resistance frequently lead to an intractable disease state in which organ failure and patient death ultimately ensue. [0252]
  • Most standard cancer therapies target cellular proliferation and rely on the differential proliferative capacities between transformed and normal cells for their efficacy. This approach is hindered by the facts that several important normal cell types are also highly proliferative and that cancer cells frequently become resistant to these agents. Thus, the therapeutic indices for traditional anti-cancer therapies rarely exceed 2.0. [0253]
  • The advent of genomics-driven molecular target identification has opened up the possibility of identifying new cancer-specific targets for therapeutic intervention that will provide safer, more effective treatments for cancer patients. Thus, newly discovered tumor-associated genes and their products can be tested for their role(s) in disease and used as tools to discover and develop innovative therapies. Genes playing important roles in any of the physiological processes outlined above can be characterized as cancer targets. [0254]
  • Genes or gene fragments identified through genomics can readily be expressed in one or more heterologous expression systems to produce functional recombinant proteins. These proteins are characterized in vitro for their biochemical properties and then used as tools in high-throughput molecular screening programs to identify chemical modulators of their biochemical activities. Activators and/or inhibitors of target protein activity can be identified in this manner and subsequently tested in cellular and in vivo disease models for anti-cancer activity. Optimization of lead compounds with iterative testing in biological models and detailed pharmacokinetic and toxicological analyses form the basis for drug development and subsequent testing in humans. [0255]
  • Protein phosphorylation is an essential component in intracellular signaling, with diverse and crucial functions including mediation of cell proliferation, survival, apoptosis, differentiation, migration and attachment. It is regulated by the balance between the opposing activities of protein kinases and protein phosphatases. Protein phosphorylation is mainly mediated by two types of protein kinases—protein tyrosine kinases and protein serine/threonine kinases. A number of protein tyrosine kinases are encoded by proto-oncogenes or viral oncogenes, and are thus strongly implicated in cancer. Protein serine/threonine kinases are known to play a role in intracellular signal transduction mediated by growth factors, cytokines, etc. inducing either cell proliferation, apoptosis or differentiation. Inhibitors of protein kinases are expected to provide efficacious therapeutic agents for the treatment of cancer. [0256]
  • CNS Disorders [0257]
  • Central and peripheral nervous system disorders also can be treated, such as primary and secondary disorders after brain injury, disorders of mood, anxiety disorders, disorders of thought and volition, disorders of sleep and wakefulness, diseases of the motor unit, such as neurogenic and myopathic disorders, neurodegenerative disorders such as Alzheimer's and Parkinson's disease, and processes of peripheral and chronic pain. [0258]
  • Pain that is associated with CNS disorders also can be treated by regulating the activity of human protein kinase-like protein. Pain which can be treated includes that associated with central nervous system disorders, such as multiple sclerosis, spinal cord injury, sciatica, failed back surgery syndrome, traumatic brain injury, epilepsy, Parkinson's disease, post-stroke, and vascular lesions in the brain and spinal cord (e.g., infarct, hemorrhage, vascular malformation). Non-central neuropathic pain includes that associated with post mastectomy pain, reflex sympathetic dystrophy (RSD), trigeminal neuralgiaradioculopathy, post-surgical pain, HIV/AIDS related pain, cancer pain, metabolic neuropathies (e.g., diabetic neuropathy, vasculitic neuropathy secondary to connective tissue disease), paraneoplastic polyneuropathy associated, for example, with carcinoma of lung, or leukemia, or lymphoma, or carcinoma of prostate, colon or stomach, trigeminal neuralgia, cranial neuralgias, and post-herpetic neuralgia. Pain associated with cancer and cancer treatment also can be treated, as can headache pain (for example, migraine with aura, migraine without aura, and other migraine disorders), episodic and chronic tension-type headache, tension-type like headache, cluster headache, and chronic paroxysmal hemicrania. [0259]
  • COPD [0260]
  • Chronic obstructive pulmonary (or airways) disease (COPD) is a condition defined physiologically as airflow obstruction that generally results from a mixture of emphysema and peripheral airway obstruction due to chronic bronchitis (Senior & Shapiro, Pulmonary Diseases and Disorders, 3d ed., New York, McGraw-Hill, 1998, pp. 659-681, 1998; Barnes, Chest 117, 10s[0261] −14S, 2000). Emphysema is characterized by destruction of alveolar walls leading to abnormal enlargement of the air spaces of the lung. Chronic bronchitis is defined clinically as the presence of chronic productive cough for three months in each of two successive years. In COPD, airflow obstruction is usually progressive and is only partially reversible. By far the most important risk factor for development of COPD is cigarette smoking, although the disease does occur in non-smokers.
  • Chronic inflammation of the airways is a key pathological feature of COPD (Senior & Shapiro, 1998). The inflammatory cell population comprises increased numbers of macrophages, neutrophils, and CD8+ lymphocytes. Inhaled irritants, such as cigarette smoke, activate macrophages which are resident in the respiratory tract, as well as epithelial cells leading to release of chemokines (e.g., interleukin-8) and other chemotactic factors. These chemotactic factors act to increase the neutrophil/-monocyte trafficking from the blood into the lung tissue and airways. Neutrophils and monocytes recruited into the airways can release a variety of potentially damaging mediators such as proteolytic enzymes and reactive oxygen species. Matrix degradation and emphysema, along with airway wall thickening, surfactant dysfunction, and mucus hypersecretion, all are potential sequelae of this inflammatory response that lead to impaired airflow and gas exchange. [0262]
  • Obesity and overweight are defined as an excess of body fat relative to lean body mass. An increase in caloric intake or a decrease in energy expenditure or both can bring about this imbalance leading to surplus energy being stored as fat. Obesity is associated with important medical morbidities and an increase in mortality. The causes of obesity are poorly understood and may be due to genetic factors, environmental factors or a combination of the two to cause a positive energy balance. In contrast, anorexia and cachexia are characterized by an imbalance in energy intake versus energy expenditure leading to a negative energy balance and weight loss. Agents that either increase energy expenditure and/or decrease energy intake, absorption or storage would be useful for treating obesity, overweight, and associated comorbidities. Agents that either increase energy intake and/or decrease energy expenditure or increase the amount of lean tissue would be useful for treating cachexia, anorexia and wasting disorders. [0263]
  • This gene, translated proteins and agents which modulate this gene or portions of the gene or its products are useful for treating obesity, overweight, anorexia, cachexia, wasting disorders, appetite suppression, appetite enhancement, increases or decreases in satiety, modulation of body weight, and/or other eating disorders such as bulimia. Also this gene, translated proteins and agents which modulate this gene or portions of the gene or its products are useful for treating obesity/overweight-associated comorbidities including hypertension, type 2 diabetes, coronary artery disease, hyper-lipidemia, stroke, gallbladder disease, gout, osteoarthritis, sleep apnea and respiratory problems, some types of cancer including endometrial, breast, prostate, and colon cancer, thrombolic disease, polycystic ovarian syndrome, reduced fertility, complications of pregnancy, menstrual irregularities, hirsutism, stress incontinence, and depression. [0264]
  • Diabetes [0265]
  • Diabetes mellitus is a common metabolic disorder characterized by an abnormal elevation in blood glucose, alterations in lipids and abnormalities (complications) in the cardiovascular system, eye, kidney and nervous system. Diabetes is divided into two separate diseases: [0266] type 1 diabetes (juvenile onset), which results from a loss of cells which make and secrete insulin, and type 2 diabetes (adult onset), which is caused by a defect in insulin secretion and a defect in insulin action.
  • [0267] Type 1 diabetes is initiated by an autoimmune reaction that attacks the insulin secreting cells (beta cells) in the pancreatic islets. Agents that prevent this reaction from occurring or that stop the reaction before destruction of the beta cells has been accomplished are potential therapies for this disease. Other agents that induce beta cell proliferation and regeneration also are potential therapies.
  • Type II diabetes is the most common of the two diabetic conditions (6% of the population). The defect in insulin secretion is an important cause of the diabetic condition and results from an inability of the beta cell to properly detect and respond to rises in blood glucose levels with insulin release. Therapies that increase the response by the beta cell to glucose would offer an important new treatment for this disease. [0268]
  • The defect in insulin action in Type II diabetic subjects is another target for therapeutic intervention. Agents that increase the activity of the insulin receptor in muscle, liver, and fat will cause a decrease in blood glucose and a normalization of plasma lipids. The receptor activity can be increased by agents that directly stimulate the receptor or that increase the intracellular signals from the receptor. Other therapies can directly activate the cellular end process, i.e. glucose transport or various enzyme systems, to generate an insulin-like effect and therefore a produce beneficial outcome. Because overweight subjects have a greater susceptibility to Type II diabetes, any agent that reduces body weight is a possible therapy. [0269]
  • Both Type I and Type diabetes can be treated with agents that mimic insulin action or that treat diabetic complications by reducing blood glucose levels. Likewise, agents that reduces new blood vessel growth can be used to treat the eye complications that develop in both diseases. [0270]
  • Cardiovascular Diseases [0271]
  • Cardiovascular diseases include the following disorders of the heart and the vascular system: congestive heart failure, myocardial infarction, ischemic diseases of the heart, all kinds of atrial and ventricular arrhythmias, hypertensive vascular diseases, and peripheral vascular diseases. [0272]
  • Heart failure is defined as a pathophysiologic state in which an abnormality of cardiac function is responsible for the failure of the heart to pump blood at a rate commensurate with the requirement of the metabolizing tissue. It includes all forms of pumping failure, such as high-output and low-output, acute and chronic, right-sided or left-sided, systolic or diastolic, independent of the underlying cause. [0273]
  • Myocardial infarction (MI) is generally caused by an abrupt decrease in coronary blood flow that follows a thrombotic occlusion of a coronary artery previously narrowed by arteriosclerosis. MI prophylaxis (primary and secondary prevention) is included, as well as the acute treatment of MI and the prevention of complications. [0274]
  • Ischemic diseases are conditions in which the coronary flow is restricted resulting in a perfusion which is inadequate to meet the myocardial requirement for oxygen. This group of diseases includes stable angina, unstable angina, and asymptomatic ischemia. [0275]
  • Arrhythmias include all forms of atrial and ventricular tachyarrhythmias (atrial tachycardia, atrial flutter, atrial fibrillation, atrio-ventricular reentrant tachycardia, preexcitation syndrome, ventricular tachycardia, ventricular flutter, and ventricular fibrillation), as well as bradycardic forms of arrhythmias. [0276]
  • Vascular diseases include primary as well as all kinds of secondary arterial hypertension (renal, endocrine, neurogenic, others). The disclosed gene and its product may be used as drug targets for the treatment of hypertension as well as for the prevention of all complications. Peripheral vascular diseases are defined as vascular diseases in which arterial and/or venous flow is reduced resulting in an imbalance between blood supply and tissue oxygen demand. It includes chronic peripheral arterial occlusive disease (PAOD), acute arterial thrombosis and embolism, inflammatory vascular disorders, Raynaud's phenomenon, and venous disorders. [0277]
  • This invention further pertains to the use of novel agents identified by the screening assays described above. Accordingly, it is within the scope of this invention to use a test compound identified as described herein in an appropriate animal model. For example, an agent identified as described herein (e.g., a modulating agent, an antisense nucleic acid molecule, a specific antibody, ribozyme, or a protein kinase-like polypeptide binding molecule) can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent. Alternatively, an agent identified as described herein can be used in an animal model to determine the mechanism of action of such an agent. Furthermore, this invention pertains to uses of novel agents identified by the above-described screening assays for treatments as described herein. [0278]
  • A reagent which affects protein kinase-like protein activity can be administered to a human cell, either in vitro or in vivo, to reduce protein kinase-like protein activity. The reagent preferably binds to an expression product of a human protein kinase-like gene. If the expression product is a protein, the reagent is preferably an antibody. For treatment of human cells ex vivo, an antibody can be added to a preparation of stem cells that have been removed from the body. The cells can then be replaced in the same or another human body, with or without clonal propagation, as is known in the art. [0279]
  • In one embodiment, the reagent is delivered using a liposome. Preferably, the liposome is stable in the animal into which it has been administered for at least about 30 minutes, more preferably for at least about 1 hour, and even more preferably for at least about 24 hours. A liposome comprises a lipid composition that is capable of targeting a reagent, particularly a polynucleotide, to a particular site in an animal, such as a human. Preferably, the lipid composition of the liposome is capable of targeting to a specific organ of an animal, such as the lung, liver, spleen, heart brain, lymph nodes, and skin. [0280]
  • A liposome useful in the present invention comprises a lipid composition that is capable of fusing with the plasma membrane of the targeted cell to deliver its contents to the cell. Preferably, the transfection efficiency of a liposome is about 0.5 μg of DNA per 16 nmole of liposome delivered to about 10[0281] 6 cells, more preferably about 1.0 μg of DNA per 16 nmole of liposome delivered to about 106 cells, and even more preferably about 2.0 μg of DNA per 16 nmol of liposome delivered to about 106 cells. Preferably, a liposome is between about 100 and 500 nm, more preferably between about 150 and 450 nm, and even more preferably between about 200 and 400 nm in diameter.
  • Suitable liposomes for use in the present invention include those liposomes standardly used in, for example, gene delivery methods known to those of skill in the art. More preferred liposomes include liposomes having a polycationic lipid composition and/or liposomes having a cholesterol backbone conjugated to polyethylene glycol. Optionally, a liposome comprises a compound capable of targeting the liposome to a particular cell type, such as a cell-specific ligand exposed on the outer surface of the liposome. [0282]
  • Complexing a liposome with a reagent such as an antisense oligonucleotide or ribozyme can be achieved using methods that are standard in the art (see, for example, U.S. Pat. No. 5,705,151). Preferably, from about 0.1 μg to about 10 μg of polynucleotide is combined with about 8 nmol of liposomes, more preferably from about 0.5 μg to about 5 μg of polynucleotides are combined with about 8 nmol liposomes, and even more preferably about 1.0 μg of polynucleotides is combined with about 8 mol liposomes. [0283]
  • In another embodiment, antibodies can be delivered to specific tissues in vivo using receptor-mediated targeted delivery. Receptor-mediated DNA delivery techniques are taught in, for example, Findeis et al. Trends in Biotechnol. 11, 202-05 (1993); Chiou et al., G[0284] ENE THERAPEUTICS: METHODS AND APPLICATIONS OF DIRECT GENE TRANSFER (J. A. Wolff, ed.) (1994); Wu & Wu, J. Biol. Chem. 263, 621-24 (1988); Wu et al., J. Biol. Chem. 269, 542-46 (1994); Zenke et al., Proc. Natl. Acad. Sci. U.S.A. 87, 3655-59 (1990); Wu et al., J. Biol. Chem. 266, 338-42 (1991).
  • Determination of a Therapeutically Effective Dose [0285]
  • The determination of a therapeutically effective dose is well within the capability of those skilled in the art. A therapeutically effective dose refers to that amount of active ingredient which increases or decreases protein kinase-like protein activity relative to the protein kinase-like protein activity which occurs in the absence of the therapeutically effective dose. [0286]
  • For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rabbits, dogs, or pigs. The animal model also can be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. [0287]
  • Therapeutic efficacy and toxicity, e.g., ED[0288] 50 (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population), can be determined by standard pharmaceutical procedures in cell cultures or experimental animals. The dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD50/ED50.
  • Pharmaceutical compositions that exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and animal studies is used in formulating a range of dosage for human use. The dosage contained in such compositions is preferably within a range of circulating concentrations that include the ED[0289] 50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
  • The exact dosage will be determined by the practitioner, in light of factors related to the subject that requires treatment. Dosage and administration are adjusted to provide sufficient levels of the active ingredient or to maintain the desired effect. Factors that can be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions can be administered every 3 to 4 days, every week, or once every two weeks depending on the half-life and clearance rate of the particular formulation. [0290]
  • Normal dosage amounts can vary from 0.1 to 100,000 micrograms, up to a total dose of about 1 g, depending upon the route of administration. Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art. Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc. [0291]
  • If the reagent is a single-chain antibody, polynucleotides encoding the antibody can be constructed and introduced into a cell either ex vivo or in vivo using well-established techniques including, but not limited to, transferrin-polycation-mediated DNA transfer, transfection with naked or encapsulated nucleic acids, liposome-mediated cellular fusion, intracellular transportation of DNA-coated latex beads, protoplast fusion, viral infection, electroporation, “gene gun,” and DEAE- or calcium phosphate-mediated transfection. [0292]
  • Effective in vivo dosages of an antibody are in the range of about 5 μg to about 50 μg/kg, about 50 μg to about 5 mg/kg, about 100 μg to about 500 μg/kg of patient body weight, and about 200 to about 250 μg/kg of patient body weight. For administration of polynucleotides encoding single-chain antibodies, effective in vivo dosages are in the range of about 100 ng to about 200 ng, 500 ng to about 50 mg, about 1 μg to about 2 mg, about 5 μg to about 500 μg, and about 20 μg to about 100 μg of DNA. [0293]
  • If the expression product is mRNA, the reagent is preferably an antisense oligo-nucleotide or a ribozyme. Polynucleotides that express antisense oligonucleotides or ribozymes can be introduced into cells by a variety of methods, as described above. [0294]
  • Preferably, a reagent reduces expression of a protein kinase-like gene or the activity of a protein kinase-like polypeptide by at least about 10, preferably about 50, more preferably about 75, 90, or 100% relative to the absence of the reagent. The effectiveness of the mechanism chosen to decrease the level of expression of a protein kinase-like gene or the activity of a protein kinase-like polypeptide can be assessed using methods well known in the art, such as hybridization of nucleotide probes to protein kinase-like protein-specific mRNA, quantitative RT-PCR, immunologic detection of a protein kinase-like polypeptide, or measurement of protein kinase-like protein activity. [0295]
  • In any of the embodiments described above, any of the pharmaceutical compositions of the invention can be administered in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy can be made by one of ordinary skill in the art, according to conventional pharmaceutical principles. The combination of therapeutic agents can act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects. [0296]
  • Any of the therapeutic methods described above can be applied to any subject in need of such therapy, including, for example, mammals such as dogs, cats, cows, horses, rabbits, monkeys, and most preferably, humans. [0297]
  • Diagnostic Methods [0298]
  • Human protein kinase-like protein also can be used in diagnostic assays for detecting diseases and abnormalities or susceptibility to diseases and abnormalities related to the presence of mutations in the nucleic acid sequences that encode the enzyme. For example, differences can be determined between the cDNA or genomic sequence encoding protein kinase-like protein in individuals afflicted with a disease and in normal individuals. If a mutation is observed in some or all of the afflicted individuals but not in normal individuals, then the mutation is likely to be the causative agent of the disease. [0299]
  • Sequence differences between a reference gene and a gene having mutations can be revealed by the direct DNA sequencing method. In addition, cloned DNA segments can be employed as probes to detect specific DNA segments. The sensitivity of this method is greatly enhanced when combined with PCR. For example, a sequencing primer can be used with a double-stranded PCR product or a single-stranded template molecule generated by a modified PCR. The sequence determination is performed by conventional procedures using radiolabeled nucleotides or by automatic sequencing procedures using fluorescent tags. [0300]
  • Genetic testing based on DNA sequence differences can be carried out by detection of alteration in electrophoretic mobility of DNA fragments in gels with or without denaturing agents. Small sequence deletions and insertions can be visualized, for example, by high resolution gel electrophoresis. DNA fragments of different sequences can be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g., Myers et al., [0301] Science 230, 1242, 1985). Sequence changes at specific locations can also be revealed by nuclease protection assays, such as RNase and S 1 protection or the chemical cleavage method (e.g., Cotton et al., Proc. Natl. Acad. Sci. USA 85, 4397-4401, 1985). Thus, the detection of a specific DNA sequence can be performed by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes and Southern blotting of genomic DNA. In addition to direct methods such as gel-electrophoresis and DNA sequencing, mutations can also be detected by in situ analysis.
  • Altered levels of protein kinase-like protein also can be detected in various tissues. Assays used to detect levels of the receptor polypeptides in a body sample, such as blood or a tissue biopsy, derived from a host are well known to those of skill in the art and include radioimmunoassays, competitive binding assays, Western blot analysis, and ELISA assays. [0302]
  • All patents and patent applications cited in this disclosure are expressly incorporated herein by reference. The above disclosure generally describes the present invention. A more complete understanding can be obtained by reference to the following specific examples, which are provided for purposes of illustration only and are not intended to limit the scope of the invention. [0303]
  • EXAMPLE 1
  • Detection of Proteine Kinase-Like Protein Activity [0304]
  • The polynucleotide of SEQ ID NO: 1 is inserted into the expression vector pCEV4 and the expression vector pCEV4-proteine kinase-like protein polypeptide obtained is transfected into human embryonic kidney 293 cells. From these cells extracts are obtained and proteine kinase-like protein activity is assayed by measuring the incorporation of 32Pi into calf thymus H1 histone from [gamma-32P]ATP. Cell extracts are incubated in a reaction mixture containing 20 mM Tris HCl at pH 7.5, 10 mM MgCl2, 20 μM ATP, 15-50 kBq of [gamma-32P]ATP, and 200 μg/ml H1 istone. The incubation is carried out for 5 min at 30° C., and the phosphorylated proteins are seperated by SDS/PAGE and visualized and quanitated by measuring the intensity of photostimulated luminescene (PSL) using a Bio-Imaging Analyzer. It is shown that the polypeptide of SEQ ID NO: 2 has a proteine kinase-like protein activity. [0305]
  • EXAMPLE 2
  • Expression of Recombinant Human Protein Kinase-Like Protein [0306]
  • The [0307] Pichia pastoris expression vector pPICZB (Invitrogen, San Diego, Calif.) is used to produce large quantities of recombinant human protein kinase-like polypeptides in yeast. The protein kinase-like protein-encoding DNA sequence is derived from SEQ ID NOS: 1, 8 and 10. Before insertion into vector pPICZB, the DNA sequence is modified by well known methods in such a way that it contains at its 5′-end an initiation codon and at its 3′-end an enterokinase cleavage site, a His6 reporter tag and a termination codon. Moreover, at both termini recognition sequences for restriction endonucleases are added and after digestion of the multiple cloning site of pPICZ B with the corresponding restriction enzymes the modified DNA sequence is ligated into pPICZB. This expression vector is designed for inducible expression in Pichia pastoris, driven by a yeast promoter. The resulting pPICZ/md-His6 vector is used to transform the yeast.
  • The yeast is cultivated under usual conditions in 5 liter shake flasks and the recombinantly produced protein isolated from the culture by affinity chromatography (Ni-NTA-Resin) in the presence of 8 M urea. The bound polypeptide is eluted with buffer, pH 3.5, and neutralized. Separation of the polypeptide from the His6 reporter tag is accomplished by site-specific proteolysis using enterokinase (Invitrogen, San Diego, Calif.) according to manufacturer's instructions. Purified human protein kinase-like polypeptide is obtained. [0308]
  • EXAMPLE 3
  • Identification of Test Compounds that Bind to Protein Kinase-Like Polypeptides [0309]
  • Purified protein kinase-like polypeptides comprising a glutathione-S-transferase protein and absorbed onto glutathione-derivatized wells of 96-well microtiter plates are contacted with test compounds from a small molecule library at pH 7.0 in a physiological buffer solution. Human protein kinase-like polypeptides comprise the amino acid sequence shown in SEQ ID NOS:2 and 9. The test compounds comprise a fluorescent tag. The samples are incubated for 5 minutes to one hour. Control samples are incubated in the absence of a test compound. [0310]
  • The buffer solution containing the test compounds is washed from the wells. Binding of a test compound to a protein kinase-like polypeptide is detected by fluorescence measurements of the contents of the wells. A test compound that increases the fluorescence in a well by at least 15% relative to fluorescence of a well in which a test compound is not incubated is identified as a compound which binds to a protein kinase-like polypeptide. [0311]
  • EXAMPLE 4
  • Identification of a Test Compound which Decreases Protein Kinase-Like Gene Expression [0312]
  • A test compound is administered to a culture of human cells transfected with a protein kinase-like protein expression construct and incubated at 37° C. for 10 to 45 minutes. A culture of the same type of cells that have not been transfected is incubated for the same time without the test compound to provide a negative control. [0313]
  • RNA is isolated from the two cultures as described in Chirgwin et al., [0314] Biochem. 18, 5294-99, 1979). Northern blots are prepared using 20 to 30 μg total RNA and hybridized with a 32P-labeled protein kinase-like protein-specific probe at 65° C. in Express-hyb (CLONTECH). The probe comprises at least 11 contiguous nucleotides selected from the complement of SEQ ID NOS: 1, 8 and 10. A test compound that decreases the protein kinase-like protein-specific signal relative to the signal obtained in the absence of the test compound is identified as an inhibitor of protein kinase-like gene expression.
  • EXAMPLE 5
  • Identification of a Test Compound which Decreases Protein Kinase-Like Protein Activity [0315]
  • A test compound is administered to a culture of human cells transfected with a protein kinase-like protein expression construct and incubated at 37° C. for 10 to 45 minutes. A culture of the same type of cells that have not been transfected is incubated for the same time without the test compound to provide a negative control. protein kinase-like protein activity is measured using the method of U.S. Pat. No. 6,194,186. [0316]
  • A test compound which decreases the protein kinase-like protein activity of the protein kinase-like protein relative to the protein kinase-like protein activity in the absence of the test compound is identified as an inhibitor of protein kinase-like protein activity. [0317]
  • EXAMPLE 6
  • Tissue-Specific Expression of Protein Kinase-Like Protein [0318]
  • The qualitative expression pattern of protein kinase-like protein in various tissues is determined by Reverse Transcription-Polymerase Chain Reaction (RT-PCR). [0319]
  • To demonstrate that protein kinase-like protein is involved in cancer, expression is determined in the following tissues: adrenal gland, bone marrow, brain, cerebellum, colon, fetal brain, fetal liver, heart, kidney, liver, lung, mammary gland, pancreas, placenta, prostate, salivary gland, skeletal muscle, small intestine, spinal cord, spleen, stomach, testis, thymus, thyroid, trachea, uterus, and peripheral blood lymphocytes. Expression in the following cancer cell lines also is determined: DU-145 (prostate), NCI-H125 (lung), HT-29 (colon), COLO-205 (colon), A-549 (lung), NCI—H460 (lung), HT-116 (colon), DLD-1 (colon), MDA-MD-231 (breast), LS 174T (colon), ZF-75 (breast), MDA-MN-435 (breast), HT-1080, MCF-7 (breast), and U87. Matched pairs of malignant and normal tissue from the same patient also are tested. [0320]
  • To demonstrate that protein kinase-like protein is involved in CNS disorders, the following tissues are screened: fetal and adult brain, muscle, heart, lung, kidney, liver, thymus, testis, colon, placenta, trachea, pancreas, kidney, gastric mucosa, colon, liver, cerebellum, skin, cortex (Alzheimer's and normal), hypothalamus, cortex, amygdala, cerebellum, hippocampus, choroid, plexus, thalamus, and spinal cord. [0321]
  • To demonstrate that protein kinase-like protein is involved in the disease process of COPD, the initial expression panel consists of RNA samples from respiratory tissues and inflammatory cells relevant to COPD: lung (adult and fetal), trachea, freshly isolated alveolar type II cells, cultured human bronchial epithelial cells, cultured small airway epithelial cells, cultured bronchial sooth muscle cells, cultured H441 cells (Clara-like), freshly isolated neutrophils and monocytes, and cultured monocytes (macrophage-like). Body map profiling also is carried out, using total RNA panels purchased from Clontech. The tissues are adrenal gland, bone marrow, brain, colon, heart, kidney, liver, lung, mammary gland, pancreas, prostate, salivary gland, skeletal muscle, small intestine, spleen, stomach, testis, thymus, trachea, thyroid, and uterus. [0322]
  • To demonstrate that protein kinase-like protein is involved in the disease process of obesity, expression is determined in the following tissues: subcutaneous adipose tissue, mesenteric adipose tissue, adrenal gland, bone marrow, brain (cerebellum, spinal cord, cerebral cortex, caudate, medulla, substantia nigra, and putamen), colon, fetal brain, heart, kidney, liver, lung, mammary gland, pancreas, placenta, prostate, salivary gland, skeletal muscle small intestine, spleen, stomach, testes, thymus, thyroid trachea, and uterus. Neuroblastoma cell lines SK-Nr-Be (2), Hr, Sk-N-As, HTB-10, IMR-32, SNSY-5Y, T3, SK-N-D2, D283, DAOY, CHP-2, U87MG, BE(2)C, T986, KANTS, M059K, CHP234, C6 (rat), SK-N-Fl, SK-PU-DW, PFSK-1, BE(2)M17, and MCIXC also are tested for protein kinase-like protein expression. As a final step, the expression of protein kinase-like protein in cells derived from normal individuals with the expression of cells derived from obese individuals is compared. [0323]
  • To demonstrate that protein kinase-like protein is involved in the disease process of diabetes, the following whole body panel is screened to show predominant or relatively high expression: subcutaneous and mesenteric adipose tissue, adrenal gland, bone marrow, brain, colon, fetal brain, heart, hypothalamus, kidney, liver, lung, mammary gland, pancreas, placenta, prostate, salivary gland, skeletal muscle, small intestine, spleen, stomach, testis, thymus, thyroid, trachea, and uterus. Human islet cells and an islet cell library also are tested. As a final step, the expression of protein kinase-like protein in cells derived from normal individuals with the expression of cells derived from diabetic individuals is compared. [0324]
  • Quantitative expression profiling. Quantitative expression profiling is performed by the form of quantitative PCR analysis called “kinetic analysis” firstly described in Higuchi et al., [0325] BioTechnology 10, 413-17, 1992, and Higuchi et al., BioTechnology 11, 1026-30, 1993. The principle is that at any given cycle within the exponential phase of PCR, the amount of product is proportional to the initial number of template copies.
  • If the amplification is performed in the presence of an internally quenched fluorescent oligonucleotide (TaqMan probe) complementary to the target sequence, the probe is cleaved by the 5′-3′ endonuclease activity of Taq DNA polymerase and a fluorescent dye released in the medium (Holland et al., [0326] Proc. Natl. Acad. Sci. U.S.A. 88, 7276-80, 1991). Because the fluorescence emission will increase in direct proportion to the amount of the specific amplified product, the exponential growth phase of PCR product can be detected and used to determine the initial template concentration (Heid et al., Genome Res. 6, 986-94, 1996, and Gibson et al., Genome Res. 6, 995-1001, 1996).
  • The amplification of an endogenous control can be performed to standardize the amount of sample RNA added to a reaction. In this kind of experiment, the control of choice is the 18S ribosomal RNA. Because reporter dyes with differing emission spectra are available, the target and the endogenous control can be independently quantified in the same tube if probes labeled with different dyes are used. [0327]
  • All “real time PCR” measurements of fluorescence are made in the ABI Prism 7700. [0328]
  • RNA extraction and cDNA preparation. Total RNA from the tissues listed above are used for expression quantification. RNAs labeled “from autopsy” were extracted from autoptic tissues with the TRIzol reagent (Life Technologies, MD) according to the manufacturer's protocol. [0329]
  • Fifty μg of each RNA were treated with DNase I for 1 hour at 37° C. in the following reaction mix: 0.2 U/μl RNase-free DNase I (Roche Diagnostics, Germany); 0.4 U/μl RNase inhibitor (PE Applied Biosystems, CA); 10 mM Tris-HCl pH 7.9; 10 mM MgCl[0330] 2; 50 mM NaCl; and 1 mM DTT.
  • After incubation, RNA is extracted once with 1 volume of phenol:chloroform:—isoamyl alcohol (24:24:1) and once with chloroform, and precipitated with {fraction (1/10)} volume of 3 M NaAcetate, pH5.2, and 2 volumes of ethanol. [0331]
  • Fifty μg of each RNA from the autoptic tissues are DNase treated with the DNA-free kit purchased from Ambion (Ambion, Tex.). After resuspension and spectrophotometric quantification, each sample is reverse transcribed with the TaqMan Reverse Transcription Reagents (PE Applied Biosystems, CA) according to the manufacturer's protocol. The final concentration of RNA in the reaction mix is 200 ng/μL. Reverse transcription is carried out with 2.5 μM of random hexamer primers. [0332]
  • TaqMan quantitative analysis. Specific primers and probe are designed according to the recommendations of PE Applied Biosystems; the probe can be labeled at the 5′ end FAM (6-carboxy-fluorescein) and at the 3′ end with TAMRA (6-carboxy-tetramethyl-rhodamine). Quantification experiments are performed on 10 ng of reverse transcribed RNA from each sample. Each determination is done in triplicate. [0333]
  • Total cDNA content is normalized with the simultaneous quantification (multiplex PCR) of the 18S ribosomal RNA using the Pre-Developed TaqMan Assay Reagents (PDAR) Control Kit (PE Applied Biosystems, CA). [0334]
  • The assay reaction mix is as follows: 1× final TaqMan Universal PCR Master Mix (from 2× stock) (PE Applied Biosystems, CA); 1×PDAR control —18S RNA (from 20× stock); 300 nM forward primer; 900 nM reverse primer; 200 nM probe; 10 ng cDNA; and water to 25 μl. [0335]
  • Each of the following steps are carried out once: pre PCR, 2 minutes at 50° C., and 10 minutes at 95° C. The following steps are carried out 40 times: denaturation, 15 seconds at 95° C., annealing/extension, 1 minute at 60° C. [0336]
  • The experiment is performed on an ABI Prism 7700 Sequence Detector (PE Applied Biosystems, CA). At the end of the run, fluorescence data acquired during PCR are processed as described in the ABI Prism 7700 user's manual in order to achieve better background subtraction as well as signal linearity with the starting target quantity. [0337]
  • EXAMPLE 7
  • Proliferation Inhibition Assay: Antisense Oligonucleotides Suppress the Growth of Cancer Cell Lines [0338]
  • The cell line used for testing is the human colon cancer cell line HCT116. Cells are cultured in RPMI-1640 with 10-15% fetal calf serum at a concentration of 10,000 cells per milliliter in a volume of 0.5 ml and kept at 37° C. in a 95% air/5% CO[0339] 2 atmosphere.
  • Phosphorothioate oligoribonucleotides are synthesized on an Applied Biosystems Model 380B DNA synthesizer using phosphoroamidite chemistry. A sequence of 24 bases complementary to the nucleotides at [0340] position 1 to 24 of SEQ ID NOS: 1, 8 and 10 is used as the test oligonucleotide. As a control, another (random) sequence is used: 5′-TCA ACT GAC TAG ATG TAC ATG GAC-3′. Following assembly and deprotection, oligonucleotides are ethanol-precipitated twice, dried, and suspended in phosphate buffered saline at the desired concentration. Purity of the oligonucleotides is tested by capillary gel electrophoresis and ion exchange HPLC. The purified oligonucleotides are added to the culture medium at a concentration of 10 μM once per day for seven days.
  • The addition of the test oligonucleotide for seven days results in significantly reduced expression of human protein kinase-like protein as determined by Western blotting. This effect is not observed with the control oligonucleotide. After 3 to 7 days, the number of cells in the cultures is counted using an automatic cell counter. The number of cells in cultures treated with the test oligonucleotide (expressed as 100%) is compared with the number of cells in cultures treated with the control oligonucleotide. The number of cells in cultures treated with the test oligonucleotide is not more than 30% of control, indicating that the inhibition of human protein kinase-like protein has an anti-proliferative effect on cancer cells. [0341]
  • EXAMPLE 8
  • In Vivo Testing of Compounds/Target Validation [0342]
  • 1. Acute Mechanistic Assays [0343]
  • 1.1. Reduction in Mitogenic Plasma Hormone Levels [0344]
  • This non-tumor assay measures the ability of a compound to reduce either the endogenous level of a circulating hormone or the level of hormone produced in response to a biologic stimulus. Rodents are administered test compound (p.o., i.p., i.v., i.m., or s.c.). At a predetermined time after administration of test compound, blood plasma is collected. Plasma is assayed for levels of the hormone of interest. If the normal circulating levels of the hormone are too low and/or variable to provide consistent results, the level of the hormone may be elevated by a pre-treatment with a biologic stimulus (i.e., LHRH may be injected i.m. into mice at a dosage of 30 ng/mouse to induce a burst of testosterone synthesis). The timing of plasma collection would be adjusted to coincide with the peak of the induced hormone response. Compound effects are compared to a vehicle-treated control group. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test. Significance is p value≦0.05 compared to the vehicle control group. [0345]
  • 1.2. Hollow Fiber Mechanism of Action Assay [0346]
  • Hollow fibers are prepared with desired cell line(s) and implanted intraperitoneally and/or subcutaneously in rodents. Compounds are administered p.o., i.p., i.v., i.m., or s.c. Fibers are harvested in accordance with specific readout assay protocol, these may include assays for gene expression (bDNA, PCR, or Taqman), or a specific biochemical activity (i.e., cAMP levels. Results are analyzed by Student's t-test or Rank Sum test after the variance between groups is compared by an F-test, with significance at p<0.05 as compared to the vehicle control group. [0347]
  • 2. Subacute Functional In Vivo Assays [0348]
  • 2.1. Reduction in Mass of Hormone Dependent Tissues [0349]
  • This is another non-tumor assay that measures the ability of a compound to reduce the mass of a hormone dependent tissue (i.e., seminal vesicles in males and uteri in females). Rodents are administered test compound (p.o., i.p., i.v., i.m., or s.c.) according to a predetermined schedule and for a predetermined duration (i.e., 1 week). At termination of the study, animals are weighed, the target organ is excised, any fluid is expressed, and the weight of the organ is recorded. Blood plasma may also be collected. Plasma may be assayed for levels of a hormone of interest or for levels of test agent. Organ weights may be directly compared or they may be normalized for the body weight of the animal. Compound effects are compared to a vehicle-treated control group. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test. Significance is p value≦0.05 compared to the vehicle control group. [0350]
  • 2.2. Hollow Fiber Proliferation Assay [0351]
  • Hollow fibers are prepared with desired cell line(s) and implanted intraperitoneally and/or subcutaneously in rodents. Compounds are administered p.o., i.p., i.v., i.m., or s.c. Fibers are harvested in accordance with specific readout assay protocol. Cell proliferation is determined by measuring a marker of cell number (i.e., MTT or LDH). The cell number and change in cell number from the starting inoculum are analyzed by Student's t-test or Rank Sum test after the variance between groups is compared by an F-test, with significance at p≦0.05 as compared to the vehicle control group. [0352]
  • 2.3 Anti-Angiogenesis Models [0353]
  • 2.3.1. Corneal Angiogenesis [0354]
  • Hydron pellets with or without growth factors or cells are implanted into a micropocket surgically created in the rodent cornea. Compound administration may be systemic or local (compound mixed with growth factors in the hydron pellet). Corneas are harvested at 7 days post implantation immediately following intracardiac infusion of colloidal carbon and are fixed in 10% formalin. Readout is qualitative scoring and/or image analysis. Qualitative scores are compared by Rank Sum test. Image analysis data is evaluated by measuring the area of neovascularization (in pixels) and group averages are compared by Student's t-test (2 tail). Significance is p<0.05 as compared to the growth factor or cells only group. [0355]
  • 2.3.2. Matrigel Angiogenesis [0356]
  • Matrigel, containing cells or growth factors, is injected subcutaneously. Compounds are administered p.o., i.p., i.v., i.m., or s.c. Matrigel plugs are harvested at predetermined time point(s) and prepared for readout. Readout is an ELISA-based assay for hemoglobin concentration and/or histological examination (i.e. vessel count, special staining for endothelial surface markers: CD31, factor-8). Readouts are analyzed by Student's t-test, after the variance between groups is compared by an F-test, with significance determined at p≦0.05 as compared to the vehicle control group. [0357]
  • 3. Primary Antitumor Efficacy [0358]
  • 3.1. Early Therapy Models [0359]
  • 3.1.1. Subcutaneous Tumor [0360]
  • Tumor cells or fragments are implanted subcutaneously on [0361] Day 0. Vehicle and/or compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule starting at a time, usually on Day 1, prior to the ability to measure the tumor burden. Body weights and tumor measurements are recorded 2-3 times weekly. Mean net body and tumor weights are calculated for each data collection day. Anti-tumor efficacy may be initially determined by comparing the size of treated (T) and control (C) tumors on a given day by a Student's t-test, after the variance between groups is compared by an F-test, with significance determined at p≦0.05. The experiment may also be continued past the end of dosing in which case tumor measurements would continue to be recorded to monitor tumor growth delay. Tumor growth delays are expressed as the difference in the median time for the treated and control groups to attain a predetermined size divided by the median time for the control group to attain that size. Growth delays are compared by generating Kaplan-Meier curves from the times for individual tumors to attain the evaluation size. Significance is p<0.05.
  • 3.1.2. Intraperitoneal/Intracranial Tumor Models [0362]
  • Tumor cells are injected intraperitoneally or intracranially on [0363] Day 0. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule starting on Day 1. Observations of morbidity and/or mortality are recorded twice daily. Body weights are measured and recorded twice weekly. Morbidity/mortality data is expressed in terms of the median time of survival and the number of long-term survivors is indicated separately. Survival times are used to generate Kaplan-Meier curves. Significance is p≦0.05 by a log-rank test compared to the control group in the experiment.
  • 3.2. Established Disease Model [0364]
  • Tumor cells or fragments are implanted subcutaneously and grown to the desired size for treatment to begin. Once at the predetermined size range, mice are randomized into treatment groups. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Tumor and body weights are measured and recorded 2-3 times weekly. Mean tumor weights of all groups over days post inoculation are graphed for comparison. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p≦0.05 as compared to the control group. Tumor measurements may be recorded after dosing has stopped to monitor tumor growth delay. Tumor growth delays are expressed as the difference in the median time for the treated and control groups to attain a predetermined size divided by the median time for the control group to attain that size. Growth delays are compared by generating Kaplan-Meier curves from the times for individual tumors to attain the evaluation size. Significance is p value≦0.05 compared to the vehicle control group. [0365]
  • 3.3. Orthotopic Disease Models [0366]
  • 3.3.1. Mammary Fat Pad Assay [0367]
  • Tumor cells or fragments, of mammary adenocarcinoma origin, are implanted directly into a surgically exposed and reflected mammary fat pad in rodents. The fat pad is placed back in its original position and the surgical site is closed. Hormones may also be administered to the rodents to support the growth of the tumors. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Tumor and body weights are measured and recorded 2-3 times weekly. Mean tumor weights of all groups over days post inoculation are graphed for comparison. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p≦0.05 as compared to the control group. [0368]
  • Tumor measurements may be recorded after dosing has stopped to monitor tumor growth delay. Tumor growth delays are expressed as the difference in the median time for the treated and control groups to attain a predetermined size divided by the median time for the control group to attain that size. Growth delays are compared by generating Kaplan-Meier curves from the times for individual tumors to attain the evaluation size. Significance is p value≦0.05 compared to the vehicle control group. In addition, this model provides an opportunity to increase the rate of spontaneous metastasis of this type of tumor. Metastasis can be assessed at termination of the study by counting the number of visible foci per target organ, or measuring the target organ weight. The means of these endpoints are compared by Student's t-test after conducting an F-test, with significance determined at p≦0.05 compared to the control group in the experiment. [0369]
  • 3.3.2. Intraprostatic Assay [0370]
  • Tumor cells or fragments, of prostatic adenocarcinoma origin, are implanted directly into a surgically exposed dorsal lobe of the prostate in rodents. The prostate is externalized through an abdominal incision so that the tumor can be implanted specifically in the dorsal lobe while verifying that the implant does not enter the seminal vesicles. The successfully inoculated prostate is replaced in the abdomen and the incisions through the abdomen and skin are closed. Hormones may also be administered to the rodents to support the growth of the tumors. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Body weights are measured and recorded 2-3 times weekly. At a predetermined time, the experiment is terminated and the animal is dissected. The size of the primary tumor is measured in three dimensions using either a caliper or an ocular micrometer attached to a dissecting scope. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p≦0.05 as compared to the control group. This model provides an opportunity to increase the rate of spontaneous metastasis of this type of tumor. Metastasis can be assessed at termination of the study by counting the number of visible foci per target organ (i.e., the lungs), or measuring the target organ weight (i.e., the regional lymph nodes). The means of these endpoints are compared by Student's t-test after conducting an F-test, with significance determined at p≦0.05 compared to the control group in the experiment. [0371]
  • 3.3.3. Intrabronchial Assay [0372]
  • Tumor cells of pulmonary origin may be implanted intrabronchially by making an incision through the skin and exposing the trachea. The trachea is pierced with the beveled end of a 25 gauge needle and the tumor cells are inoculated into the main bronchus using a flat-ended 27 gauge needle with a 90° bend. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Body weights are measured and recorded 2-3 times weekly. At a predetermined time, the experiment is terminated and the animal is dissected. The size of the primary tumor is measured in three dimensions using either a caliper or an ocular micrometer attached to a dissecting scope. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p≦0.05 as compared to the control group. This model provides an opportunity to increase the rate of spontaneous metastasis of this type of tumor. Metastasis can be assessed at termination of the study by counting the number of visible foci per target organ (i.e., the contralateral lung), or measuring the target organ weight. The means of these endpoints are compared by Student's t-test after conducting an F-test, with significance determined at p<0.05 compared to the control group in the experiment. [0373]
  • 3.3.4. Intracecal Assay [0374]
  • Tumor cells of gastrointestinal origin may be implanted intracecally by making an abdominal incision through the skin and externalizing the intestine. Tumor cells are inoculated into the cecal wall without penetrating the lumen of the intestine using a 27 or 30 gauge needle. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Body weights are measured and recorded 2-3 times weekly. At a predetermined time, the experiment is terminated and the animal is dissected. The size of the primary tumor is measured in three dimensions using either a caliper or an ocular micrometer attached to a dissecting scope. An F-test is preformed to determine if the variance is equal or unequal followed by a Student's t-test to compare tumor sizes in the treated and control groups at the end of treatment. Significance is p≦0.05 as compared to the control group. This model provides an opportunity to increase the rate of spontaneous metastasis of this type of tumor. Metastasis can be assessed at termination of the study by counting the number of visible foci per target organ (i.e., the liver), or measuring the target organ weight. The means of these endpoints are compared by Student's t-test after conducting an F-test, with significance determined at p≦0.05 compared to the control group in the experiment. [0375]
  • 4. Secondary (Metastatic) Antitumor Efficacy [0376]
  • 4.1. Spontaneous Metastasis [0377]
  • Tumor cells are inoculated s.c. and the tumors allowed to grow to a predetermined range for spontaneous metastasis studies to the lung or liver. [0378]
  • These primary tumors are then excised. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule which may include the period leading up to the excision of the primary tumor to evaluate therapies directed at inhibiting the early stages of tumor metastasis. Observations of morbidity and/or mortality are recorded daily. Body weights are measured and recorded twice weekly. Potential endpoints include survival time, numbers of visible foci per target organ, or target organ weight. When survival time is used as the endpoint the other values are not determined. Survival data is used to generate Kaplan-Meier curves. Significance is p≦0.05 by a log-rank test compared to the control group in the experiment. The mean number of visible tumor foci, as determined under a dissecting microscope, and the mean target organ weights are compared by Student's t-test after conducting an F-test, with significance determined at p≦0.05 compared to the control group in the experiment for both of these endpoints. [0379]
  • 4.2. Forced Metastasis [0380]
  • Tumor cells are injected into the tail vein, portal vein, or the left ventricle of the heart in experimental (forced) lung, liver, and bone metastasis studies, respectively. Compounds are administered p.o., i.p., i.v., i.m., or s.c. according to a predetermined schedule. Observations of morbidity and/or mortality are recorded daily. Body weights are measured and recorded twice weekly. Potential endpoints include survival time, numbers of visible foci per target organ, or target organ weight. When survival time is used as the endpoint the other values are not determined. Survival data is used to generate Kaplan-Meier curves. Significance is p≦0.05 by a log-rank test compared to the control group in the experiment. The mean number of visible tumor foci, as determined under a dissecting microscope, and the mean target organ weights are compared by Student's t-test after conducting an F-test, with significance at p≦0.05 compared to the vehicle control group in the experiment for both endpoints. [0381]
  • EXAMPLE 9
  • In Vivo Testing of Compounds/Target Validation [0382]
  • 1. Pain [0383]
  • Acute Pain [0384]
  • Acute pain is measured on a hot plate mainly in rats. Two variants of hot plate testing are used: In the classical variant animals are put on a hot surface (52 to 56° C.) and the latency time is measured until the animals show nocifensive behavior, such as stepping or foot licking. The other variant is an increasing temperature hot plate where the experimental animals are put on a surface of neutral temperature. Subsequently this surface is slowly but constantly heated until the animals begin to lick a hind paw. The temperature which is reached when hind paw licking begins is a measure for pain threshold. [0385]
  • Compounds are tested against a vehicle treated control group. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to pain testing. [0386]
  • Persistent Pain [0387]
  • Persistent pain is measured with the formalin or capsaicin test, mainly in rats. A solution of 1 to 5% formalin or 10 to 100 μg capsaicin is injected into one hind paw of the experimental animal. After formalin or capsaicin application the animals show nocifensive reactions like flinching, licking and biting of the affected paw. The number of nocifensive reactions within a time frame of up to 90 minutes is a measure for intensity of pain. [0388]
  • Compounds are tested against a vehicle treated control group. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to formalin or capsaicin administration. [0389]
  • Neuropathic Pain [0390]
  • Neuropathic pain is induced by different variants of unilateral sciatic nerve injury mainly in rats. The operation is performed under anesthesia. The first variant of sciatic nerve injury is produced by placing loosely constrictive ligatures around the common sciatic nerve. The second variant is the tight ligation of about the half of the diameter of the common sciatic nerve. In the next variant, a group of models is used in which tight ligations or transections are made of either the L5 and L6 spinal nerves, or the L % spinal nerve only. The fourth variant involves an axotomy of two of the three terminal branches of the sciatic nerve (tibial and common peroneal nerves) leaving the remaining sural nerve intact whereas the last variant comprises the axotomy of only the tibial branch leaving the sural and common nerves uninjured. Control animals are treated with a sham operation. [0391]
  • Postoperatively, the nerve injured animals develop a chronic mechanical allodynia, cold allodynioa, as well as a thermal hyperalgesia. Mechanical allodynia is measured by means of a pressure transducer (electronic von Frey Anesthesiometer, IITC Inc.-Life Science Instruments, Woodland Hills, SA, USA; Electronic von Frey System, Somedic Sales AB, Hörby, Sweden). Thermal hyperalgesia is measured by means of a radiant heat source (Plantar Test, Ugo Basile, Comerio, Italy), or by means of a cold plate of 5 to 10° C. where the nocifensive reactions of the affected hind paw are counted as a measure of pain intensity. A further test for cold induced pain is the counting of nocifensive reactions, or duration of nocifensive responses after plantar administration of acetone to the affected hind limb. Chronic pain in general is assessed by registering the circadanian rhythms in activity (Suijo and Arndt, Universitat zu Köln, Cologne, Germany), and by scoring differences in gait (foot print patterns; FOOTPRINTS program, Klapdor et al., 1997. A low cost method to analyze footprint patterns. J. Neurosci. Methods 75, 49-54). [0392]
  • Compounds are tested against sham operated and vehicle treated control groups. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to pain testing. [0393]
  • Inflammatory Pain [0394]
  • Inflammatory pain is induced mainly in rats by injection of 0.75 mg carrageenan or complete Freund's adjuvant into one hind paw. The animals develop an edema with mechanical allodynia as well as thermal hyperalgesia. [0395]
  • Mechanical allodynia is measured by means of a pressure transducer (electronic von Frey Anesthesiometer, IITC Inc.-Life Science Instruments, Woodland Hills, SA, USA). Thermal hyperalgesia is measured by means of a radiant heat source (Plantar Test, Ugo Basile, Comerio, Italy, Paw thermal stimulator, G. Ozaki, University of California, USA). For edema measurement two methods are being used. In the first method, the animals are sacrificed and the affected hindpaws sectioned and weighed. The second method comprises differences in paw volume by measuring water displacement in a plethysmometer (Ugo Basile, Comerio, Italy). [0396]
  • Compounds are tested against uninflamed as well as vehicle treated control groups. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to pain testing. [0397]
  • Diabetic Neuropathic Pain [0398]
  • Rats treated with a single intraperitoneal injection of 50 to 80 mg/kg streptozotocin develop a profound hyperglycemia and mechanical allodynia within 1 to 3 weeks. Mechanical allodynia is measured by means of a pressure transducer (electronic von Frey Anesthesiometer, IITC Inc.-Life Science Instruments, Woodland Hills, SA, USA). [0399]
  • Compounds are tested against diabetic and non-diabetic vehicle treated control groups. Substance application is performed at different time points via different application routes (i.v., i.p., p.o., i.t., i.c.v., s.c., intradermal, transdermal) prior to pain testing. [0400]
  • 2. Parkinson's Disease [0401]
  • 6-Hydroxydopamine (6-OH-DA) Lesion [0402]
  • Degeneration of the dopaminergic nigrostriatal and striatopallidal pathways is the central pathological event in Parkinson's disease. This disorder has been mimicked experimentally in rats using single/sequential unilateral stereotaxic injections of 6-OH-DA into the medium forebrain bundle (MFB). [0403]
  • Male Wistar rats (Harlan Winkelmann, Germany), weighing 200±250 g at the beginning of the experiment, are used. The rats are maintained in a temperature- and humidity-controlled environment under a 12 h light/dark cycle with free access to food and water when not in experimental sessions. The following in vivo protocols are approved by the governmental authorities. All efforts are made to minimize animal suffering, to reduce the number of animals used, and to utilize alternatives to in vivo techniques. [0404]
  • Animals are administered pargyline on the day of surgery (Sigma, St. Louis, Mo., USA; 50 mg/kg i.p.) in order to inhibit metabolism of 6-OHDA by monoamine oxidase and desmethylimipramine HCl (Sigma; 25 mg/kg i.p.) in order to prevent uptake of 6-OHDA by noradrenergic terminals. Thirty minutes later the rats are anesthetized with sodium pentobarbital (50 mg/kg) and placed in a stereotaxic frame. In order to lesion the DA nigrostriatal pathway 4 μl of 0.01% ascorbic acid-saline containing 8 μg of 6-OHDA HBr (Sigma) are injected into the left medial fore-brain bundle at a rate of 1 μl/min (2.4 mm anterior, 1.49 mm lateral, −2.7 mm ventral to Bregma and the skull surface). The needle is left in place an additional 5 min to allow diffusion to occur. [0405]
  • Stepping Test [0406]
  • Forelimb akinesia is assessed three weeks following lesion placement using a modified stepping test protocol. In brief, the animals are held by the experimenter with one hand fixing the hindlimbs and slightly raising the hind part above the surface. One paw is touching the table, and is then moved slowly sideways (5 s for 1 m), first in the forehand and then in the backhand direction. The number of adjusting steps is counted for both paws in the backhand and forehand direction of movement. The sequence of testing is right paw forehand and backhand adjusting stepping, followed by left paw forehand and backhand directions. The test is repeated three times on three consecutive days, after an initial training period of three days prior to the first testing. Forehand adjusted stepping reveals no consistent differences between lesioned and healthy control animals. Analysis is therefore restricted to backhand adjusted stepping. [0407]
  • Balance Test [0408]
  • Balance adjustments following postural challenge are also measured during the stepping test sessions. The rats are held in the same position as described in the stepping test and, instead of being moved sideways, tilted by the experimenter towards the side of the paw touching the table. This maneuver results in loss of balance and the ability of the rats to regain balance by forelimb movements is scored on a scale ranging from 0 to 3. [0409] Score 0 is given for a normal forelimb placement. When the forelimb movement is delayed but recovery of postural balance detected, score 1 is given. Score 2 represents a clear, yet insufficient, forelimb reaction, as evidenced by muscle contraction, but lack of success in recovering balance, and score 3 is given for no reaction of movement. The test is repeated three times a day on each side for three consecutive days after an initial training period of three days prior to the first testing.
  • Staircase Test (Paw Reaching) [0410]
  • A modified version of the staircase test is used for evaluation of paw reaching behavior three weeks following primary and secondary lesion placement. Plexiglass test boxes with a central platform and a removable staircase on each side are used. The apparatus is designed such that only the paw on the same side at each staircase can be used, thus providing a measure of independent forelimb use. For each test the animals are left in the test boxes for 15 min. The double staircase is filled with 7×3 chow pellets (Precision food pellets, formula: P, purified rodent diet, size 45 mg; Sandown Scientific) on each side. After each test the number of pellets eaten (successfully retrieved pellets) and the number of pellets taken (touched but dropped) for each paw and the success rate (pellets eaten/pellets taken) are counted separately. After three days of food deprivation (12 g per animal per day) the animals are tested for 11 days. Full analysis is conducted only for the last five days. [0411]
  • MPTP Treatment [0412]
  • The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine (MPTP) causes degeneration of mesencephalic dopaminergic (DAergic) neurons in rodents, non-human primates, and humans and, in so doing, reproduces many of the symptoms of Parkinson's disease. MPTP leads to a marked decrease in the levels of dopamine and its metabolites, and in the number of dopaminergic terminals in the striatum as well as severe loss of the tyrosine hydroxylase (TH)-immunoreactive cell bodies in the substantia nigra, pars compacta. [0413]
  • In order to obtain severe and long-lasting lesions, and to reduce mortality, animals receive single injections of MPTP, and are then tested for severity of lesion 7-10 days later. Successive MPTP injections are administered on [0414] days 1, 2 and 3. Animals receive application of 4 mg/kg MPTP hydrochloride (Sigma) in saline once daily. All injections are intraperitoneal (i.p.) and the MPTP stock solution is frozen between injections. Animals are decapitated on day 11.
  • Immunohistology [0415]
  • At the completion of behavioral experiments, all animals are anaesthetized with 3 ml thiopental (1 g/40 ml i.p., Tyrol Pharma). The mice are perfused transcardially with 0.01 M PBS (pH 7.4) for 2 min, followed by 4% paraformaldehyde (Merck) in PBS for 15 min. The brains are removed and placed in 4% paraformaldehyde for 24 h at 4° C. For dehydration they are then transferred to a 20% sucrose (Merck) solution in 0.1 M PBS at 4° C. until they sink. The brains are frozen in methylbutan at −20° C. for 2 min and stored at −70° C. Using a sledge microtome (mod. 3800-Frigocut, Leica), 25 μm sections are taken from the genu of the corpus callosum (AP 1.7 mm) to the hippocarnpus (AP 21.8 mm) and from AP 24.16 to AP 26.72. Forty-six sections are cut and stored in assorters in 0.25 M Tris buffer (pH 7.4) for immunohistochemistry. [0416]
  • A series of sections is processed for free-floating tyrosine hydroxylase (TH) immunohistochemistry. Following three rinses in 0.1 M PBS, endogenous peroxidase activity is quenched for 10 min in 0.3% H[0417] 2O2+PBS. After rinsing in PBS, sections are preincubated in 10% normal bovine serum (Sigma) for 5 min as blocking agent and transferred to either primary anti-rat TH rabbit antiserum (dilution 1:2000).
  • Following overnight incubation at room temperature, sections for TH immunoreactivity are rinsed in PBS (2×10 min) and incubated in biotinylated anti-rabbit immunoglobulin G raised in goat (dilution 1:200) (Vector) for 90 min, rinsed repeatedly and transferred to Vectastain ABC (Vector) solution for 1 h. 3,.3′-Diaminobenzidine tetrahydrochloride (DAB; Sigma) in 0.1 M PBS, supplemented with 0.005% H[0418] 2O2, serves as chromogen in the subsequent visualization reaction. Sections are mounted on to gelatin-coated slides, left to dry overnight, counter-stained with hematoxylin dehydrated in ascending alcohol concentrations and cleared in butylacetate. Coverslips are mounted on entellan.
  • Rotarod Test [0419]
  • We use a modification of the procedure described by Rozas and Labandeira-Garcia (1997), with a CR-1 Rotamex system (Columbus Instruments, Columbus, Ohio) comprising an IBM-compatible personal computer, a CIO-24 data acquisition card, a control unit, and a four-lane rotarod unit. The rotarod unit consists of a rotating spindle (diameter 7.3 cm) and individual compartments for each mouse. The system software allows preprogramming of session protocols with varying rotational speeds (0-80 rpm). Infrared beams are used to detect when a mouse has fallen onto the base grid beneath the rotarod. The system logs the fall as the end of the experiment for that mouse, and the total time on the rotarod, as well as the time of the fall and all the set-up parameters, are recorded. The system also allows a weak current to be passed through the base grid, to aid training. [0420]
  • 3. Dementia [0421]
  • The Object Recognition Task [0422]
  • The object recognition task has been designed to assess the effects of experimental manipulations on the cognitive performance of rodents. A rat is placed in an open field, in which two identical objects are present. The rats inspects both objects during the first trial of the object recognition task. In a second trial, after a retention interval of for example 24 hours, one of the two objects used in the first trial, the ‘familiar’ object, and a novel object are placed in the open field. The inspection time at each of the objects is registered. The basic measures in the OR task is the time spent by a rat exploring the two object the second trial. Good retention is reflected by higher exploration times towards the novel than the ‘familiar’ object. [0423]
  • Administration of the putative cognition enhancer prior to the first trial predominantly allows assessment of the effects on acquisition, and eventually on consolidation processes. Administration of the testing compound after the first trial allows to assess the effects on consolidation processes, whereas administration before the second trial allows to measure effects on retrieval processes. [0424]
  • The Passive Avoidance Task [0425]
  • The passive avoidance task assesses memory performance in rats and mice. The inhibitory avoidance apparatus consists of a two-compartment box with a light compartment and a dark compartment. The two compartments are separated by a guillotine door that can be operated by the experimenter. A threshold of 2 cm separates the two compartments when the guillotine door is raised. When the door is open, the illumination in the dark compartment is about 2 lux. The light intensity is about 500 lux at the center of the floor of the light compartment. [0426]
  • Two habituation sessions, one shock session, and a retention session are given, separated by inter-session intervals of 24 hours. In the habituation sessions and the retention session the rat is allowed to explore the apparatus for 300 sec. The rat is placed in the light compartment, facing the wall opposite to the guillotine door. After an accommodation period of 15 sec. the guillotine door is opened so that all parts of the apparatus can be visited freely. Rats normally avoid brightly lit areas and will enter the dark compartment within a few seconds. [0427]
  • In the shock session the guillotine door between the compartments is lowered as soon as the rat has entered the dark compartment with its four paws, and a scrambled 1 mA footshock is administered for 2 sec. The rat is removed from the apparatus and put back into its home cage. The procedure during the retention session is identical to that of the habituation sessions. [0428]
  • The step-through latency, that is the first latency of entering the dark compartment (in sec.) during the retention session is an index of the memory performance of the animal; the longer the latency to enter the dark compartment, the better the retention is. A testing compound in given half an hour before the shock session, together with 1 mg*kg[0429] −1 scopolamine. Scopolamine impairs the memory performance during the retention session 24 hours later. If the test compound increases the enter latency compared with the scopolamine-treated controls, is likely to possess cognition enhancing potential.
  • The Morris Water Escape Task [0430]
  • The Morris water escape task measures spatial orientation learning in rodents. It is a test system that has extensively been used to investigate the effects of putative therapeutic on the cognitive functions of rats and mice. The performance of an animal is assessed in a circular water tank with an escape platform that is submerged about 1 cm below the surface of the water. The escape platform is not visible for an animal swimming in the water tank. Abundant extra-maze cues are provided by the furniture in the room, including desks, computer equipment, a second water tank, the presence of the experimenter, and by a radio on a shelf that is playing softly. [0431]
  • The animals receive four trials during five daily acquisition sessions. A trial is started by placing an animal into the pool, facing the wall of the tank. Each of four starting positions in the quadrants north, east, south, and west is used once in a series of four trials; their order is randomized. The escape platform is always in the same position. A trial is terminated as soon as the animal had climbs onto the escape platform or when 90 seconds have elapsed, whichever event occurs first. The animal is allowed to stay on the platform for 30 seconds. Then it is taken from the platform and the next trial is started. If an animal did not find the platform within 90 seconds it is put on the platform by the experimenter and is allowed to stay there for 30 seconds. After the fourth trial of the fifth daily session, an additional trial is given as a probe trial: the platform is removed, and the time the animal spends in the four quadrants is measured for 30 or 60 seconds. In the probe trial, all animals start from the same start position, opposite to the quadrant where the escape platform had been positioned during acquisition. [0432]
  • Four different measures are taken to evaluate the performance of an animal during acquisition training: escape latency, traveled distance, distance to platform, and swimming speed. The following measures are evaluated for the probe trial: time (s) in quadrants and traveled distance (cm) in the four quadrants. The probe trial provides additional information about how well an animal learned the position of the escape platform. If an animal spends more time and swims a longer distance in the quadrant where the platform had been positioned during the acquisition sessions than in any other quadrant, one concludes that the platform position has been learned well. [0433]
  • In order to assess the effects of putative cognition enhancing compounds, rats or mice with specific brain lesions which impair cognitive functions, or animals treated with compounds such as scopolamine or MK-801, which interfere with normal learning, or aged animals which suffer from cognitive deficits, are used. [0434]
  • The T-Maze Spontaneous Alternation Task [0435]
  • The T-maze spontaneous alternation task (TeMCAT) assesses the spatial memory performance in mice. The start arm and the two goal arms of the T-maze are provided with guillotine doors which can be operated manually by the experimenter. A mouse is put into the start arm at the beginning of training. The guillotine door is closed. In the first trial, the ‘forced trial’, either the left or right goal arm is blocked by lowering the guillotine door. After the mouse has been released from the start arm, it will negotiate the maze, eventually enter the open goal arm, and return to the start position, where it will be confined for 5 seconds, by lowering the guillotine door. Then, the animal can choose freely between the left and right goal arm (all guillotine-doors opened) during 14 ‘free choice’ trials. As soon a the mouse has entered one goal arm, the other one is closed. The mouse eventually returns to the start arm and is free to visit whichever go alarm it wants after having been confined to the start arm for 5 seconds. After completion of 14 free choice trials in one session, the animal is removed from the maze. During training, the animal is never handled. [0436]
  • The percent alternations out of 14 trials is calculated. This percentage and the total time needed to complete the first forced trial and the subsequent 14 free choice trials (in s) is analyzed. Cognitive deficits are usually induced by an injection of scopolamine, 30 min before the start of the training session. Scopolamine reduced the per-cent alternations to chance level, or below. A cognition enhancer, which is always administered before the training session, will at least partially, antagonize the scopolamine-induced reduction in the spontaneous alternation rate. [0437]
  • EXAMPLE 10
  • Expression of Human Protein Kinase-Like Protein [0438]
  • Total RNA used for Taqman quantitative analysis were either purchased (Clontech,CA) or extracted from tissues using TRIzol reagent (Life Technologies, MD) according to a modified vendor protocol which utilizes the Rneasy protocol (Qiagen, CA). [0439]
  • One hundred μg of each RNA were treated with DNase I using RNase free- DNase (Qiagen, CA) for use with RNeasy or QiaAmp columns. [0440]
  • After elution and quantitation with Ribogreen (Molecular Probes Inc., OR), each sample was reverse transcribed using the GibcoBRL Superscript II First Strand Synthesis System for RT-PCR according to vendor protocol (Life Technologies, MD). The final concentration of RNA in the reaction mix was 50 ng/μL. Reverse transcription was performed with 50 ng of random hexamers. [0441]
  • Specific primers and probe were designed according to PE Applied Biosystems' Primer Express program recommendations and are listed below: [0442]
    forward primer: 5′-(CTGTGACCTGAAGTCGGACAAC)-3′
    reverse primer: 5′-(GGAATGACTGCCTCGAAATCC)-3′
    probe: SYBR Green
  • Quantitation experiments were performed on 25 ng of reverse transcribed RNA from each sample. 18S ribosomal RNA was measured as a control using the Pre-Developed TaqMan Assay Reagents (PDAR)(PE Applied Biosystems, CA). The assay reaction mix was as follows: [0443]
    final
    TaqMan SYBR Green PCR Master Mix (2x)
    (PE Applied Biosystems, CA)
    Forward primer 300 nM
    Reverse primer 300 nM
    cDNA  25 ng
    Water to  25 uL
  • PCR conditions: [0444]
    Once:  2′ minutes at 50° C.
    10 minutes at 95° C.
    40cycles: 15 sec. at 95° C.
     1 minute at 60° C.
  • The experiment was performed on an ABI Prism 7700 Sequence Detector (PE Applied Biosystems, CA). At the end of the run, fluorescence data acquired during PCR were processed as described in the ABI Prism 7700 user's manual. Fold change was calculated using the delta-delta CT method with normalization to the 18S values. Relative expression was calculated by normalizing to 18s (D Ct), then making the highest expressing [0445] tissue 100 and everything else relative to it. Copy number conversion was performed without normalization using the formula Cn=10(Ct-40.007)/-3.623.
  • The results are shown in FIGS. 13, 14, [0446] 15 and 16.
  • Protein kinase-like protein expression in adipose and skeletal muscle could be regulated to increase insulin sensitivity. [0447]
  • REFERENCES
  • 1. Protein kinases 6. The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification. FASEB J 1995 May;9(8):576-96 [0448]
  • 2. Protein kinase catalytic domain sequence database: identification of conserved features of primary structure and classification of family members. Methods Enzymol 1991;200:38-62 [0449]
  • 1 12 1 858 DNA Homo sapiens 1 atgaccgact tcccggccag gctcttcctg gagaacagca agctggagca cagcgaggac 60 gagggcagcg tcctgggcca gggcggcagt ggcaccgtca tctaccgggc ccggtaccag 120 ggccagcctg tggccgtcaa gcgcttccac atcaaaaaat tcaagaactt tgctaacgta 180 ccggcagaca ccatgctgag gcacctgcgg gccaccgatg ccatgaagaa cttctccgag 240 ttccggcagg aggccagcat gctgcacgcg ctgcagcacc cctgcatcgt ggcgctcatc 300 ggcatcagca tccacccgct ctgcttcgcc ctggagctcg cgccgctcag cagcctcaac 360 accgtgctgt ccgagaacgc cagagattct tcctttatac ccctgggaca catgctcacc 420 caaaaaatag cctaccagat cgcctcgggc ctggcctacc tgcacaagaa aaacatcatc 480 ttctgtgacc tgaagtcgga caacattctg gtgtggtccc ttgacgtcaa ggagcacatc 540 aacatcaagc tatctgacta cgggatttcg aggcagtcat tccatgaggg cgccctaggc 600 gtggagggca ctcctggcta ccaggcccca gagatcaggc ctcgcattgt atatgatgag 660 aaggtagata tgttctccta tggaatggtg ctctacgagt tgctgtcagg acagcgccct 720 gcactgggcc accaccagct ccagattgcc aagaagctgt ccaagggcat ccgcccggtt 780 ctggggcagc cggaggaagt gcagttccgg cgactgcagg cgctcatgat ggagtgctgg 840 gacactaagc cagagaag 858 2 286 PRT Homo sapiens 2 Met Thr Asp Phe Pro Ala Arg Leu Phe Leu Glu Asn Ser Lys Leu Glu 1 5 10 15 His Ser Glu Asp Glu Gly Ser Val Leu Gly Gln Gly Gly Ser Gly Thr 20 25 30 Val Ile Tyr Arg Ala Arg Tyr Gln Gly Gln Pro Val Ala Val Lys Arg 35 40 45 Phe His Ile Lys Lys Phe Lys Asn Phe Ala Asn Val Pro Ala Asp Thr 50 55 60 Met Leu Arg His Leu Arg Ala Thr Asp Ala Met Lys Asn Phe Ser Glu 65 70 75 80 Phe Arg Gln Glu Ala Ser Met Leu His Ala Leu Gln His Pro Cys Ile 85 90 95 Val Ala Leu Ile Gly Ile Ser Ile His Pro Leu Cys Phe Ala Leu Glu 100 105 110 Leu Ala Pro Leu Ser Ser Leu Asn Thr Val Leu Ser Glu Asn Ala Arg 115 120 125 Asp Ser Ser Phe Ile Pro Leu Gly His Met Leu Thr Gln Lys Ile Ala 130 135 140 Tyr Gln Ile Ala Ser Gly Leu Ala Tyr Leu His Lys Lys Asn Ile Ile 145 150 155 160 Phe Cys Asp Leu Lys Ser Asp Asn Ile Leu Val Trp Ser Leu Asp Val 165 170 175 Lys Glu His Ile Asn Ile Lys Leu Ser Asp Tyr Gly Ile Ser Arg Gln 180 185 190 Ser Phe His Glu Gly Ala Leu Gly Val Glu Gly Thr Pro Gly Tyr Gln 195 200 205 Ala Pro Glu Ile Arg Pro Arg Ile Val Tyr Asp Glu Lys Val Asp Met 210 215 220 Phe Ser Tyr Gly Met Val Leu Tyr Glu Leu Leu Ser Gly Gln Arg Pro 225 230 235 240 Ala Leu Gly His His Gln Leu Gln Ile Ala Lys Lys Leu Ser Lys Gly 245 250 255 Ile Arg Pro Val Leu Gly Gln Pro Glu Glu Val Gln Phe Arg Arg Leu 260 265 270 Gln Ala Leu Met Met Glu Cys Trp Asp Thr Lys Pro Glu Lys 275 280 285 3 394 PRT Homo sapiens 3 Ala Glu Leu Thr Leu Glu Glu Ile Ile Gly Ile Gly Gly Phe Gly Lys 1 5 10 15 Val Tyr Arg Ala Phe Trp Ile Gly Asp Glu Val Ala Val Lys Ala Ala 20 25 30 Arg His Asp Pro Asp Glu Asp Ile Ser Gln Thr Ile Glu Asn Val Arg 35 40 45 Gln Glu Ala Lys Leu Phe Ala Met Leu Lys His Pro Asn Ile Ile Ala 50 55 60 Leu Arg Gly Val Cys Leu Lys Glu Pro Asn Leu Cys Leu Val Met Glu 65 70 75 80 Phe Ala Arg Gly Gly Pro Leu Asn Arg Val Leu Ser Gly Lys Arg Ile 85 90 95 Pro Pro Asp Ile Leu Val Asn Trp Ala Val Gln Ile Ala Arg Gly Met 100 105 110 Asn Tyr Leu His Asp Glu Ala Ile Val Pro Ile Ile His Arg Asp Leu 115 120 125 Lys Ser Ser Asn Ile Leu Ile Leu Gln Lys Val Glu Asn Gly Asp Leu 130 135 140 Ser Asn Lys Ile Leu Lys Ile Thr Asp Phe Gly Leu Ala Arg Glu Trp 145 150 155 160 His Arg Thr Thr Lys Met Ser Ala Ala Gly Thr Tyr Ala Trp Met Ala 165 170 175 Pro Glu Val Ile Arg Ala Ser Met Phe Ser Lys Gly Ser Asp Val Trp 180 185 190 Ser Tyr Gly Val Leu Leu Trp Glu Leu Leu Thr Gly Glu Val Pro Phe 195 200 205 Arg Gly Ile Asp Gly Leu Arg Val Ala Tyr Gly Val Ala Met Asn Lys 210 215 220 Leu Ala Leu Pro Ile Pro Ser Thr Cys Pro Glu Pro Phe Ala Lys Leu 225 230 235 240 Met Glu Asp Cys Trp Asn Pro Asp Pro His Ser Arg Pro Ser Phe Thr 245 250 255 Asn Ile Leu Asp Gln Leu Thr Thr Ile Glu Glu Ser Gly Phe Phe Glu 260 265 270 Met Pro Lys Asp Ser Phe His Cys Leu Gln Asp Asn Trp Lys His Glu 275 280 285 Ile Gln Glu Met Phe Asp Gln Leu Arg Ala Lys Glu Lys Glu Leu Arg 290 295 300 Thr Trp Glu Glu Glu Leu Thr Arg Ala Ala Leu Gln Gln Lys Asn Gln 305 310 315 320 Glu Glu Leu Leu Arg Arg Arg Glu Gln Glu Leu Ala Glu Arg Glu Ile 325 330 335 Asp Ile Leu Glu Arg Glu Leu Asn Ile Ile Ile His Gln Leu Cys Gln 340 345 350 Glu Lys Pro Arg Val Lys Lys Arg Lys Gly Lys Phe Arg Lys Ser Arg 355 360 365 Leu Ala Gln Pro Val Leu Pro Phe Pro His Gly His Ser Arg Cys Pro 370 375 380 Gly Gly Thr Gly Ser Ser Trp Gly Gly Gln 385 390 4 473 DNA Homo sapiens 4 cacgaggctg gtccctgaac tgttcatgac cgacttcccg gccaggctct tcctggagaa 60 cagcaagctg gagcacagcg aggacgaggg cagcgtcctg ggccagggcg gcagtggcac 120 cgtcatctac cgggcccggt accagggcca gcctgtggcc gtcaagcgct tccacatcaa 180 aaaattcaag aactttgcta acgtaccggc agacaccatg ctgaggcacc tgcgggccac 240 cgatgccatg aagaacttct ccgagttccg gcaggaggcc agcatgctgc acgcgctgca 300 gcacccctgc atcgtggcgc tcatcggcat cagcatccac ccgctctgct tcgccctgga 360 gctcgcgccg ctcagcagcc tcaacaccgt gctgtccgag aacgccagag attcttcctt 420 tatacccctg ggacacatgc tcacccaaaa aatagcctac cagatcgcct cgg 473 5 454 DNA Homo sapiens 5 cacgaggctg gtccctgaac tgttcatgac cgacttcccg gccaggctct tcctggagaa 60 cagcaagctg gagcacagcg aggacgaggg cagcgtcctg ggccagggcg gcagtggcac 120 cgtcatctac cgggcccggt accagggcca gcctgtggcc gtcaagcgct tccacatcaa 180 aaaattcaag aactttgcta acgtaccggc agacaccatg ctgaggcacc tgcgggccac 240 cgatgccatg aagaacttct ccgagttccg gcaggaggcc agcatgctgc acgcgctgca 300 gcacccctgc atcgtggcgc tcatcggcat cagcatccac ccgctctgct tcgccctgga 360 gctcgcgccg ctcagcagcc tcaacaccgt gctgtccgag aacgccagag attcttcctt 420 tatacccctg ggacacatgc tcacccaaaa aata 454 6 1066 DNA Homo sapiens 6 ctcttcctgt gagaacagca agctggagca cagcgaggac gagggcagcg tcctgggcca 60 cggcggcagt ggcaccgtca tctaccgggc ccggtaccag ggccagcctg tggccgtcaa 120 gcgcttccac atcaaaaaat tcaagaactt tgctaacgta ccggcagaca ccatgctgag 180 gcacctgcgg gccaccgatg ccatgaagaa cttctccgag ttccggcagg aggccagcat 240 gctgcacgcg ctgcagcacc cctgcatcgt ggcgctcatc ggcatcagca tccacccgac 300 tctgcttcgc cctggagctc gcgccgctca gcagcctcaa caccgtgctg tccgagaacg 360 ccagagattc ttcctttata cccctgggac acatgctcac ccaaaaaata gcctaccaga 420 tcgcctcggg cctggcctac ctgcacaaga aaaacatcat cttctgtgac ctgaagtcgg 480 acaacattct ggtgtggtcc cttgacgtca aggagcacat caacatcaag ctatctgact 540 acgggatttc gaggcagtca ttccatgagg gcgccctagg cgtggagggc actcctggct 600 accaggcccc agagatcagg cctcgcattg tatatgatga gaaggtagca tatgtctcct 660 atggaatggt gctctacgag ttgctgtcag gacaggggcc tgcattgggc caccaacagc 720 ttccgattgc caagaggtgt ccaaggcatc cgcccggtac tgaggacagc cggaaggaac 780 tgcaatccgg cgatgcatgg ctcattattg agatgctggg aaactataat caagcgaacg 840 gagcactgtg gccctatacg taggtagccc caattagacg accctcacat tatgaaccaa 900 tcggtaataa acgtgtacgt gcgggactag cagccaccta taacacacag ccacggccac 960 ctgggctgtg atgcatacaa gacgctcaaa cacgcgggtc acagaaagcc tcatcgagtc 1020 gcatccaccc gagagagtag cgccgccgaa ctagtataga cggtat 1066 7 471 DNA Homo sapiens misc_feature (452)..(452) n=a, c, g or t 7 cacgaggaaa aaattcaaga actttgctaa cgtaccggca gacaccatgc tgaggcacct 60 gcgggccacc gatgccatga agaacttctc cgagttccgg caggaggcca gcatgctgca 120 cgcgctgcag cacccctgca tcgtggcgct catcggcatc agcatccacc cgctctgctt 180 cgccctggag ctcgcgccgc tcagcagcct caacaccgtg ctgtccgaga acgccagaga 240 ttcttccttt atacccctgg gacacatgct cacccaaaaa atagcctacc agatcgcctc 300 gggcctggcc tacctgcaca agaaaaacat catcttctgt gacctgaagt cggacaacat 360 tctggtgtgg tcccttgacg tcaaggagca catcaacatc aagctatctg actacgggat 420 ttcgaggcag tcattccatg agggcgccct angcgtggag ggcactcctg g 471 8 5475 DNA Homo sapiens 8 ccccttaatg attttgcaca ggccccaaag caatgctgtc ttacctgcgt gctcagctgc 60 ggaaagcgga aaagtgcaag ctgatgaaga tgatcatcgt gggtcccccg cgccagggca 120 agtccaccct cctggagatc ttacagacgg ggagggcccc ccaggtggtg catggagagg 180 ccaccatcag gaccaccaag tgggagctcc agaggccggc tggctcaaga gccaaggttg 240 agtccgtgga gttcaacgtc tgggacatcg ggggaccggc cagcatggcc actgtcaacc 300 agtgcttctt cacggacaag gccctgtacg tggtggtctg gaacctggcg ctgggggagg 360 aggccgtggc caacctccag ttctggctgc tcaacatcga ggccaaggcc ccaaacgccg 420 tggtgctggt ggtcgggacg cacctggatt taattgaagc caagttccgt gtggaaagga 480 ttgcaacgct gcgtgcctat gtgctggcac tctgccgctc cccctccggc tccagggcca 540 caggcttccc agacatcacc ttcaaacact tacatgagat ttcctgcaag agcctggaag 600 gtcaggaagg gctgcgacag ctgattttcc acgtcacgtg cagcatgaag gacgtgggca 660 gcaccatcgg ctgccagcga ctggcagggc ggctgatccc caggagctac ctgagcctgc 720 aggaggccgt gctggcagag cagcagcgcc gcagccggga cgacgacgtg cagtacctga 780 cggacaggca gctggagcag ctggtggagc agacgcccga caacgacatc aaggactacg 840 aggacctgca gtcagccatc agcttcctca tagaaaccgg caccctgctc catttcccgg 900 acaccagcca cggcctgagg aacctctact tcctcgaccc tatttggctc tccgaatgtc 960 tgcagaggat ctttaatatt aagggctctc ggtcagtggc caagaatggg gtgatcagag 1020 cagaagacct caggatgctg ctggtgggga ctggcttcac gcagcagacg gaagagcagt 1080 acttccagtt cctggccaag tttgagatcg ccctgcccgt cgccaatgac agctacctcc 1140 tgccccatct ccttccatct aaacctggcc tggacaccca cggtatgcgg caccccacag 1200 ccaacaccat tcagagggta tttaagatga gcttcgttcc cgttggcttc tggcaaaggt 1260 ttatagcacg gatgctgatc agcctggcgg agatggacct gcagcttttt gaaaacaaga 1320 agaatactaa aagcaggaac aggaaagtca ccatttacag ttttacagga aaccagagaa 1380 atcgctgtag cacattcaga gtgaaaagaa atcagaccat ctattggcag gaagggctcc 1440 tggtcacttt tgatgggggc tacctcagtg tggaatcttc cgacgtgaac tggaaaaaga 1500 agaaaagcgg aggaatgaaa attgtttgcc aatcagaagt gagggacttc tcagccatgg 1560 ctttcatcac ggaccacgtc aattccttga ttgatcagtg gtttcccgcc ctgacggcca 1620 cagagagcga cgggacgcca ctcatggagc agtacgtgcc ctgcccggtc tgcgagacag 1680 cctgggccca gcacacggac cccagtgaga aatcagagga tgtgcagtac ttcgacatgg 1740 aagactgtgt cctgacggcc atcgagcggg acttcatctc ctgccccaga cacccggacc 1800 tccccgtgcc gctgcaggag ctggtccctg aactgttcat gaccgacttc ccggccaggc 1860 tcttcctgga gaacagcaag ctggagcaca gcgaggacga gggcagcgtc ctgggccagg 1920 gcggcagtgg caccgtcatc taccgggccc ggtaccaggg ccagcctgtg gccgtcaagc 1980 gcttccacat caaaaaattc aagaactttg ctaacgtacc ggcagacacc atgctgaggc 2040 acctgcgggc caccgatgcc atgaagaact tctccgagtt ccggcaggag gccagcatgc 2100 tgcacgcgct gcagcacccc tgcatcgtgg cgctcatcgg catcagcatc cacccgctct 2160 gcttcgccct ggagctcgcg ccgctcagca gcctcaacac cgtgctgtcc gagaacgcca 2220 gagattcttc ctttataccc ctgggacaca tgctcaccca aaaaatagcc taccagatcg 2280 cctcgggcct ggcctacctg cacaagaaaa acatcatctt ctgtgacctg aagtcggaca 2340 acattctggt gtggtccctt gacgtcaagg agcacatcaa catcaagcta tctgactacg 2400 ggatttcgag gcagtcattc catgagggcg ccctaggcgt ggagggcact cctggctacc 2460 aggccccaga gatcaggcct cgcattgtat atgatgagaa ggtagatatg ttctcctatg 2520 gaatggtgct ctacgagttg ctgtcaggac agcgccctgc actgggccac caccagctcc 2580 agattgccaa gaagctgtcc aagggcatcc gcccggttct ggggcagccg gaggaagtgc 2640 agttccggcg actgcaggcg ctcatgatgg agtgctggga cactaagcca gagaagcgac 2700 cgctggccct gtcggtggtg agccagatga aggacccgac ttttgccacc ttcatgtatg 2760 aactgtgctg tgggaagcag acagccttct tctcatccca gggccaggag tacaccgtgg 2820 tgttttggga tggaaaagag gagtccagga actacacggt ggtgaacaca gagaagggcc 2880 tcatggaggt gcagaggatg tgctgccctg ggatgaaggt gagctgccag ctccaggtcc 2940 agagatccct gtggacagcc accgaggacc agaaaatcta catctacacc ctcaagggca 3000 tgtgcccctt aaacacaccc caacaggcct tggatactcc agctgtcgtc acctgcttct 3060 tggccgtgcc tgttattaaa aagaattcct acctggtctt agcgggcctc gccgatgggc 3120 ttgtggctgt gtttcccgtg gtgcggggca ccccaaagga cagctgctcc tacctgtgct 3180 cacacacagc caacaggtcc aagttcagca tcgcggatga agacgcacgg cagaacccct 3240 acccagtgaa ggccatggag gtggtcaaca gcggctctga ggtctggtac agcaatgggc 3300 cgggcctcct tgtcatcgac tgtgcctccc tggagatctg caggcggctg gagccctaca 3360 tggccccctc catggttacg tcagtcgtgt gcagctctga gggcagaggg gaggaggtcg 3420 tctggtgcct ggatgacaag gccaactcct tggtgatgta ccactccacc acctaccagc 3480 tgtgtgcccg gtacttctgc ggggtcccca gccccctcag ggacatgttt cccgtgcggc 3540 ccttggacac ggaacccccg gcagccagcc acacggccaa cccaaaggtg cctgaggggg 3600 actccatcgc ggacgtgagc atcatgtaca gtgaggagct gggcacgcag atcctgatcc 3660 accaggaatc actcactgac tactgctcca tgtcctccta ctcctcatcc ccaccccgcc 3720 aggctgccag gtccccctca agcctcccca gctccccagc aagttcttcc agtgtgcctt 3780 tctccaccga ctgcgaggac tcagacatgc tacatacgcc cggtgctgcc tccgacaggt 3840 ctgagcatga cctgaccccc atggacgggg agaccttcag ccagcacctg caggccgtga 3900 agatcctcgc cgtcagagac ctcatttggg tccccaggcg cggtggagat gttatcgtca 3960 ttggcctgga gaaggattct gaagcccagc ggggccgagt cattgccgtc ttaaaagccc 4020 gagagctgac tccgcatggg gtgctggtgg atgctgccgt ggtggcaaag gacactgttg 4080 tgtgcacctt tgaaaatgaa aacacagagt ggtgcctggc cgtctggagg ggctggggcg 4140 ccagggagtt cgacattttc taccagtcct acgaggagct gggccggctg gaggcttgca 4200 ctcgcaagag aaggtaattc ctgtggaatg actgtcacac atcagagctg gctggcccgg 4260 ggctgcagcc tgacccctct gccatcggcc tctagttctc caaggaccta gaagacagat 4320 ggagttctcc cctgaactcc ttgctgctaa gaagtgctga gaagttactc gcctggcggt 4380 ggctccaggg ttctctggtt ctctggagca gagttctctg aataccccat cccccaactg 4440 ctgattttac agccccaggg aagacagtgg tatcaggctg ggagcggcct cctctggcct 4500 cccccatcag tttgcaggag caggggtgca ggatcctgtt ctgagctggg tcaaacaaag 4560 cagggccggg ccttcctgcc atccccaggt ctcagatgga attacactag aggccctccg 4620 ctgggaagca cttgaggtag ggcaggaggg gggctgtgac ccctgccctt tccccgccag 4680 agacctcggg ctctcagcac attccacagg ctcctgagtc cccgaggcct gggccagctt 4740 gggcaagcca agatcagatg tctctgtgtt cgggaaggtc tccgtgtggg aaagcccttg 4800 ggggatcccg ggtgaggagt gttgccccat ccagagaatg aatgagttcc tttaagtgcc 4860 accgccagca agcccagagg cacacagtcc gagtgcaccc gcttagcctt tacattcctc 4920 tccaccgaca aaaggaaggg gaaactcaat cagcaggact tcagaaaggg ccttgtgttt 4980 atagctttgt caagtaaatt tggacgcagc tggagcacag gccctgtttg tttgcacata 5040 ataatcttgt ttatcacttt aaaaaattca gtaatatctc agcagtcagg cttctggttg 5100 tgaaatcaca ttgtatggga tttataccaa attatgtatt tgctaaacat tcactgcaca 5160 cgtgtacagc ggagtacgaa aaggaacgtt gtccacaggg gatttatgga tacaacagca 5220 aacattttat aaactatgca catgcattac acacatgcac acacatatgc acacacatgt 5280 gcaaacatag ccactttttt gtcaagagtt accctttggg gctccttaaa ccagaatggg 5340 agtttgaaag agagatcata ctccagctga agtttgttga cccttttcta aaattaaaaa 5400 gatcaaattt agtatttgct ggatatgcag ggagatgaga ctcttttaat ctcaaaataa 5460 acagattctt tcaag 5475 9 1394 PRT Homo sapiens 9 Met Leu Ser Tyr Leu Arg Ala Gln Leu Arg Lys Ala Glu Lys Cys Lys 1 5 10 15 Leu Met Lys Met Ile Ile Val Gly Pro Pro Arg Gln Gly Lys Ser Thr 20 25 30 Leu Leu Glu Ile Leu Gln Thr Gly Arg Ala Pro Gln Val Val His Gly 35 40 45 Glu Ala Thr Ile Arg Thr Thr Lys Trp Glu Leu Gln Arg Pro Ala Gly 50 55 60 Ser Arg Ala Lys Val Glu Ser Val Glu Phe Asn Val Trp Asp Ile Gly 65 70 75 80 Gly Pro Ala Ser Met Ala Thr Val Asn Gln Cys Phe Phe Thr Asp Lys 85 90 95 Ala Leu Tyr Val Val Val Trp Asn Leu Ala Leu Gly Glu Glu Ala Val 100 105 110 Ala Asn Leu Gln Phe Trp Leu Leu Asn Ile Glu Ala Lys Ala Pro Asn 115 120 125 Ala Val Val Leu Val Val Gly Thr His Leu Asp Leu Ile Glu Ala Lys 130 135 140 Phe Arg Val Glu Arg Ile Ala Thr Leu Arg Ala Tyr Val Leu Ala Leu 145 150 155 160 Cys Arg Ser Pro Ser Gly Ser Arg Ala Thr Gly Phe Pro Asp Ile Thr 165 170 175 Phe Lys His Leu His Glu Ile Ser Cys Lys Ser Leu Glu Gly Gln Glu 180 185 190 Gly Leu Arg Gln Leu Ile Phe His Val Thr Cys Ser Met Lys Asp Val 195 200 205 Gly Ser Thr Ile Gly Cys Gln Arg Leu Ala Gly Arg Leu Ile Pro Arg 210 215 220 Ser Tyr Leu Ser Leu Gln Glu Ala Val Leu Ala Glu Gln Gln Arg Arg 225 230 235 240 Ser Arg Asp Asp Asp Val Gln Tyr Leu Thr Asp Arg Gln Leu Glu Gln 245 250 255 Leu Val Glu Gln Thr Pro Asp Asn Asp Ile Lys Asp Tyr Glu Asp Leu 260 265 270 Gln Ser Ala Ile Ser Phe Leu Ile Glu Thr Gly Thr Leu Leu His Phe 275 280 285 Pro Asp Thr Ser His Gly Leu Arg Asn Leu Tyr Phe Leu Asp Pro Ile 290 295 300 Trp Leu Ser Glu Cys Leu Gln Arg Ile Phe Asn Ile Lys Gly Ser Arg 305 310 315 320 Ser Val Ala Lys Asn Gly Val Ile Arg Ala Glu Asp Leu Arg Met Leu 325 330 335 Leu Val Gly Thr Gly Phe Thr Gln Gln Thr Glu Glu Gln Tyr Phe Gln 340 345 350 Phe Leu Ala Lys Phe Glu Ile Ala Leu Pro Val Ala Asn Asp Ser Tyr 355 360 365 Leu Leu Pro His Leu Leu Pro Ser Lys Pro Gly Leu Asp Thr His Gly 370 375 380 Met Arg His Pro Thr Ala Asn Thr Ile Gln Arg Val Phe Lys Met Ser 385 390 395 400 Phe Val Pro Val Gly Phe Trp Gln Arg Phe Ile Ala Arg Met Leu Ile 405 410 415 Ser Leu Ala Glu Met Asp Leu Gln Leu Phe Glu Asn Lys Lys Asn Thr 420 425 430 Lys Ser Arg Asn Arg Lys Val Thr Ile Tyr Ser Phe Thr Gly Asn Gln 435 440 445 Arg Asn Arg Cys Ser Thr Phe Arg Val Lys Arg Asn Gln Thr Ile Tyr 450 455 460 Trp Gln Glu Gly Leu Leu Val Thr Phe Asp Gly Gly Tyr Leu Ser Val 465 470 475 480 Glu Ser Ser Asp Val Asn Trp Lys Lys Lys Lys Ser Gly Gly Met Lys 485 490 495 Ile Val Cys Gln Ser Glu Val Arg Asp Phe Ser Ala Met Ala Phe Ile 500 505 510 Thr Asp His Val Asn Ser Leu Ile Asp Gln Trp Phe Pro Ala Leu Thr 515 520 525 Ala Thr Glu Ser Asp Gly Thr Pro Leu Met Glu Gln Tyr Val Pro Cys 530 535 540 Pro Val Cys Glu Thr Ala Trp Ala Gln His Thr Asp Pro Ser Glu Lys 545 550 555 560 Ser Glu Asp Val Gln Tyr Phe Asp Met Glu Asp Cys Val Leu Thr Ala 565 570 575 Ile Glu Arg Asp Phe Ile Ser Cys Pro Arg His Pro Asp Leu Pro Val 580 585 590 Pro Leu Gln Glu Leu Val Pro Glu Leu Phe Met Thr Asp Phe Pro Ala 595 600 605 Arg Leu Phe Leu Glu Asn Ser Lys Leu Glu His Ser Glu Asp Glu Gly 610 615 620 Ser Val Leu Gly Gln Gly Gly Ser Gly Thr Val Ile Tyr Arg Ala Arg 625 630 635 640 Tyr Gln Gly Gln Pro Val Ala Val Lys Arg Phe His Ile Lys Lys Phe 645 650 655 Lys Asn Phe Ala Asn Val Pro Ala Asp Thr Met Leu Arg His Leu Arg 660 665 670 Ala Thr Asp Ala Met Lys Asn Phe Ser Glu Phe Arg Gln Glu Ala Ser 675 680 685 Met Leu His Ala Leu Gln His Pro Cys Ile Val Ala Leu Ile Gly Ile 690 695 700 Ser Ile His Pro Leu Cys Phe Ala Leu Glu Leu Ala Pro Leu Ser Ser 705 710 715 720 Leu Asn Thr Val Leu Ser Glu Asn Ala Arg Asp Ser Ser Phe Ile Pro 725 730 735 Leu Gly His Met Leu Thr Gln Lys Ile Ala Tyr Gln Ile Ala Ser Gly 740 745 750 Leu Ala Tyr Leu His Lys Lys Asn Ile Ile Phe Cys Asp Leu Lys Ser 755 760 765 Asp Asn Ile Leu Val Trp Ser Leu Asp Val Lys Glu His Ile Asn Ile 770 775 780 Lys Leu Ser Asp Tyr Gly Ile Ser Arg Gln Ser Phe His Glu Gly Ala 785 790 795 800 Leu Gly Val Glu Gly Thr Pro Gly Tyr Gln Ala Pro Glu Ile Arg Pro 805 810 815 Arg Ile Val Tyr Asp Glu Lys Val Asp Met Phe Ser Tyr Gly Met Val 820 825 830 Leu Tyr Glu Leu Leu Ser Gly Gln Arg Pro Ala Leu Gly His His Gln 835 840 845 Leu Gln Ile Ala Lys Lys Leu Ser Lys Gly Ile Arg Pro Val Leu Gly 850 855 860 Gln Pro Glu Glu Val Gln Phe Arg Arg Leu Gln Ala Leu Met Met Glu 865 870 875 880 Cys Trp Asp Thr Lys Pro Glu Lys Arg Pro Leu Ala Leu Ser Val Val 885 890 895 Ser Gln Met Lys Asp Pro Thr Phe Ala Thr Phe Met Tyr Glu Leu Cys 900 905 910 Cys Gly Lys Gln Thr Ala Phe Phe Ser Ser Gln Gly Gln Glu Tyr Thr 915 920 925 Val Val Phe Trp Asp Gly Lys Glu Glu Ser Arg Asn Tyr Thr Val Val 930 935 940 Asn Thr Glu Lys Gly Leu Met Glu Val Gln Arg Met Cys Cys Pro Gly 945 950 955 960 Met Lys Val Ser Cys Gln Leu Gln Val Gln Arg Ser Leu Trp Thr Ala 965 970 975 Thr Glu Asp Gln Lys Ile Tyr Ile Tyr Thr Leu Lys Gly Met Cys Pro 980 985 990 Leu Asn Thr Pro Gln Gln Ala Leu Asp Thr Pro Ala Val Val Thr Cys 995 1000 1005 Phe Leu Ala Val Pro Val Ile Lys Lys Asn Ser Tyr Leu Val Leu 1010 1015 1020 Ala Gly Leu Ala Asp Gly Leu Val Ala Val Phe Pro Val Val Arg 1025 1030 1035 Gly Thr Pro Lys Asp Ser Cys Ser Tyr Leu Cys Ser His Thr Ala 1040 1045 1050 Asn Arg Ser Lys Phe Ser Ile Ala Asp Glu Asp Ala Arg Gln Asn 1055 1060 1065 Pro Tyr Pro Val Lys Ala Met Glu Val Val Asn Ser Gly Ser Glu 1070 1075 1080 Val Trp Tyr Ser Asn Gly Pro Gly Leu Leu Val Ile Asp Cys Ala 1085 1090 1095 Ser Leu Glu Ile Cys Arg Arg Leu Glu Pro Tyr Met Ala Pro Ser 1100 1105 1110 Met Val Thr Ser Val Val Cys Ser Ser Glu Gly Arg Gly Glu Glu 1115 1120 1125 Val Val Trp Cys Leu Asp Asp Lys Ala Asn Ser Leu Val Met Tyr 1130 1135 1140 His Ser Thr Thr Tyr Gln Leu Cys Ala Arg Tyr Phe Cys Gly Val 1145 1150 1155 Pro Ser Pro Leu Arg Asp Met Phe Pro Val Arg Pro Leu Asp Thr 1160 1165 1170 Glu Pro Pro Ala Ala Ser His Thr Ala Asn Pro Lys Val Pro Glu 1175 1180 1185 Gly Asp Ser Ile Ala Asp Val Ser Ile Met Tyr Ser Glu Glu Leu 1190 1195 1200 Gly Thr Gln Ile Leu Ile His Gln Glu Ser Leu Thr Asp Tyr Cys 1205 1210 1215 Ser Met Ser Ser Tyr Ser Ser Ser Pro Pro Arg Gln Ala Ala Arg 1220 1225 1230 Ser Pro Ser Ser Leu Pro Ser Ser Pro Ala Ser Ser Ser Ser Val 1235 1240 1245 Pro Phe Ser Thr Asp Cys Glu Asp Ser Asp Met Leu His Thr Pro 1250 1255 1260 Gly Ala Ala Ser Asp Arg Ser Glu His Asp Leu Thr Pro Met Asp 1265 1270 1275 Gly Glu Thr Phe Ser Gln His Leu Gln Ala Val Lys Ile Leu Ala 1280 1285 1290 Val Arg Asp Leu Ile Trp Val Pro Arg Arg Gly Gly Asp Val Ile 1295 1300 1305 Val Ile Gly Leu Glu Lys Asp Ser Glu Ala Gln Arg Gly Arg Val 1310 1315 1320 Ile Ala Val Leu Lys Ala Arg Glu Leu Thr Pro His Gly Val Leu 1325 1330 1335 Val Asp Ala Ala Val Val Ala Lys Asp Thr Val Val Cys Thr Phe 1340 1345 1350 Glu Asn Glu Asn Thr Glu Trp Cys Leu Ala Val Trp Arg Gly Trp 1355 1360 1365 Gly Ala Arg Glu Phe Asp Ile Phe Tyr Gln Ser Tyr Glu Glu Leu 1370 1375 1380 Gly Arg Leu Glu Ala Cys Thr Arg Lys Arg Arg 1385 1390 10 4216 DNA Homo sapiens 10 caaagcaatg ctgtcttacc tgcgtgctca gctgcggaaa gcggaaaagt gcaagctgat 60 gaagatgatc atcgtgggtc ccccgcgcca gggcaagtcc accctcctgg agatcttaca 120 gacggggagg gccccccagg tggtgcatgg agaggccacc atcaggacca ccaagtggga 180 gctccagagg ccggctggct caagagccaa ggttgagtcc gtggagttca acgtctggga 240 catcggggga ccggccagca tggccactgt caaccagtgc ttcttcacgg acaaggccct 300 gtacgtggtg gtctggaacc tggcgctggg ggaggaggcc gtggccaacc tccagttctg 360 gctgctcaac atcgaggcca aggccccaaa cgccgtggtg ctggtggtcg ggacgcacct 420 ggatttaatt gaagccaagt tccgtgtgga aaggattgca acgctgcgtg cctatgtgct 480 ggcactctgc cgctccccct ccggctccag ggccacaggc ttcccagaca tcaccttcaa 540 acacttacat gagatttcct gcaagagcct ggaaggtcag gaagggctgc gacagctgat 600 tttccacgtc acgtgcagca tgaaggacgt gggcagcacc atcggctgcc agcgactggc 660 agggcggctg atccccagga gctacctgag cctgcaggag gccgtgctgg cagagcagca 720 gcgccgcagc cgggacgacg acgtgcagta cctgacggac aggcagctgg agcagctggt 780 ggagcagacg cccgacaacg acatcaagga ctacgaggac ctgcagtcag ccatcagctt 840 cctcatagaa accggcaccc tgctccattt cccggacacc agccacggcc tgaggaacct 900 ctacttcctc gaccctattt ggctctccga atgtctgcag aggatcttta atattaaggg 960 ctctcggtca gtggccaaga atggggtgat cagagcagaa gacctcagga tgctgctggt 1020 ggggactggc ttcacgcagc agacggaaga gcagtacttc cagttcctgg ccaagtttga 1080 gatcgccctg cccgtcgcca atgacagcta cctcctgccc catctccttc catctaaacc 1140 tggcctggac acccacggta tgcggcaccc cacagccaac accattcaga gggtatttaa 1200 gatgagcttc gttcccgttg gcttctggca aaggtttata gcacggatgc tgatcagcct 1260 ggcggagatg gacctgcagc tttttgaaaa caagaagaat actaaaagca ggaacaggaa 1320 agtcaccatt tacagtttta caggaaacca gagaaatcgc tgtagcacat tcagagtgaa 1380 aagaaatcag accatctatt ggcaggaagg gctcctggtc acttttgatg ggggctacct 1440 cagtgtggaa tcttccgacg tgaactggaa aaagaagaaa agcggaggaa tgaaaattgt 1500 ttgccaatca gaagtgaggg acttctcagc catggctttc atcacggacc acgtcaattc 1560 cttgattgat cagtggtttc ccgccctgac agccacagag agcgacggga cgccactcat 1620 ggagcagtac gtgccctgcc cggtctgcga gacagcctgg gcccagcaca cggaccccag 1680 tgagaaatca gaggatgtgc agtacttcga catggaagac tgtgtcctga cggccatcga 1740 gcgggacttc atctcctgcc ccagacaccc ggacctcccc gtgccgctgc aggagctggt 1800 ccctgaactg ttcatgaccg acttcccggc caggctcttc ctggagaaca gcaagctgga 1860 gcacagcgag gacgagggca gcgtcctggg ccagggcggc agtggcaccg tcatctaccg 1920 ggcccggtac cagggccagc ctgtggccgt caagcgcttc cacatcaaaa aattcaagaa 1980 ctttgctaac gtaccggcag acaccatgct gaggcacctg cgggccaccg atgccatgaa 2040 gaacttctcc gagttccggc aggaggccag catgctgcac gcgctgcagc acccctgcat 2100 cgtggcgctc atcggcatca gcatccaccc gctctgcttc gccctggagc tcgcgccgct 2160 cagcagcctc aacaccgtgc tgtccgagaa cgccagagat tcttccttta tacccctggg 2220 acacatgctc acccaaaaaa tagcctacca gatcgcctcg ggcctggcct acctgcacaa 2280 gaaaaacatc atcttctgtg acctgaagtc ggacaacatt ctggtgtggt cccttgacgt 2340 caaggagcac atcaacatca agctatctga ctacgggatt tcgaggcagt cattccatga 2400 gggcgcccta ggcgtggagg gcactcctgg ctaccaggcc ccagagatca ggcctcgcat 2460 tgtatatgat gagaaggtag atatgttctc ctatggaatg gtgctctacg agttgctgtc 2520 aggacagcgc cctgcactgg gccaccacca gctccagatt gccaagaagc tgtccaaggg 2580 catccgcccg gttctggggc agccggagga agtgcagttc cggcgactgc aggcgctcat 2640 gatggagtgc tgggacacta agccagagaa gcgaccgctg gccctgtcgg tggtgagcca 2700 gatgaaggac ccgacttttg ccaccttcat gtatgaactg tgctgtggga agcagacagc 2760 cttcttctca tcccagggcc aggagtacac cgtggtgttt tgggatggaa aagaggagtc 2820 caggaactac acggtggtga acacagagaa gggcctcatg gaggtgcaga ggatgtgctg 2880 ccctgggatg aaggtgagct gccagctcca ggtccagaga tccctgtgga cagccaccga 2940 ggaccagaaa atctacatct acaccctcaa gggcatgtgc cccttaaaca caccccaaca 3000 ggccttggat actccagctg tcgtcacctg cttcttggcc gtgcctgtta ttaaaaagaa 3060 ttcctacctg gtcttagcgg gcctcgccga tgggcttgtg gctgtgtttc ccgtggtgcg 3120 gggcacccca aaggacagct gctcctacct gtgctcacac acagccaaca ggtccaagtt 3180 cagcatcgcg gatgaagacg cacggcagaa cccctaccca gtgaaggcca tggaggtggt 3240 caacagcggc tctgaggtct ggtacagcaa tgggccgggc ctccttgtca tcgactgtgc 3300 ctccctggag atctgcaggc ggctggagcc ctacatggcc ccctccatgg ttacgtcagt 3360 cgtgtgcagc tctgagggca gaggggagga ggtcgtctgg tgcctggatg acaaggccaa 3420 ctccttggtg atgtaccact ccaccaccta ccagctgtgt gcccggtact tctgcggggt 3480 ccccagcccc ctcagggaca tgtttcccgt gcggcccttg gacacggaac ccccggcagc 3540 cagccacacg gccaacccaa aggtgcctga gggggactcc atcgcggacg tgagcatcat 3600 gtacagtgag gagctgggca cgcagatcct gatccaccag gaatcactca ctgactactg 3660 ctccatgtcc tcctactcct catccccacc ccgccaggct gccaggtccc cctcaagcct 3720 ccccagctcc ccagcaagtt cttccagtgt gcctttctcc accgactgcg aggactcaga 3780 catgctacat acgcccggtg ctgcctccga caggtctgag catgacctga cccccatgga 3840 cggggagacc ttcagccagc acctgcaggc cgtgaagatc ctcgccgtca gagacctcat 3900 ttgggtcccc aggcgcggtg gagatgttat cgtcattggc ctggagaagg attctgaagc 3960 ccagcggggc cgagtcattg ccgtcttaaa agcccgagag ctgactccgc atggggtgct 4020 ggtggatgct gccgtggtgg caaaggacac tgttgtgtgc acctttgaaa atgaaaacac 4080 agagtggtgc ctggccgtct ggaggggctg gggcgccagg gagttcgaca ttttctacca 4140 gtcctacgag gagctgggcc ggctggaggc ttgcactcgc aagagaaggt aattcctgtg 4200 gaatgactgt cacaca 4216 11 22 DNA Artificial misc_feature forward primer 11 ctgtgacctg aagtcggaca ac 22 12 21 DNA Homo sapiens misc_feature reverse primer 12 ggaatgactg cctcgaaatc c 21

Claims (71)

1. An isolated polynucleotide being selected from the group consisting of:
a) a polynucleotide encoding a proteine kinase-like protein polypeptide comprising an amino acid sequence selected form the group consisting of:
amino acid sequences which are at least about 35% identical to the amino acid sequence shown in SEQ ID NO: 2;
the amino acid sequence shown in SEQ ID NO: 2;
amino acid sequences which are at least about 35% identical to the amino acid sequence shown in SEQ ID NO: 9; and
the amino acid sequence shown in SEQ ID NO: 9.
b) a polynucleotide comprising the sequence of SEQ ID NOS: 1; 8 or 10.
c) a polynucleotide which hybridizes under stringent conditions to a polynucleotide specified in (a) and (b) and encodes a proteine kinase-like protein polypeptide;
d) a polynucleotide the sequence of which deviates from the polynucleotide sequences specified in (a) to (c) due to the degeneration of the genetic code and encodes a proteine kinase-like protein polypeptide; and
e) a polynucleotide which represents a fragment, derivative or allelic variation of a polynucleotide sequence specified in (a) to (d) and encodes a proteine kinase-like protein polypeptide.
2. An expression vector containing any polynucleotide of claim 1.
3. A host cell containing the expression vector of claim 2.
4. A substantially purified proteine kinase-like protein polypeptide encoded by a polynucleotide of claim 1.
5. A method for producing a proteine kinase-like protein polypeptide, wherein the method comprises the following steps:
a) culturing the host cell of claim 3 under conditions suitable for the expression of the proteine kinase-like protein polypeptide; and
b) recovering the proteine kinase-like protein polypeptide from the host cell culture.
6. A method for detection of a polynucleotide encoding a proteine kinase-like protein polypeptide in a biological sample comprising the following steps:
a) hybridizing any polynucleotide of claim 1 to a nucleic acid material of a biological sample, thereby forming a hybridization complex; and
b) detecting said hybridization complex.
7. The method of claim 6, wherein before hybridization, the nucleic acid material of the biological sample is amplified.
8. A method for the detection of a polynucleotide of claim 1 or a proteine kinase-like protein polypeptide of claim 4 comprising the steps of:
contacting a biological sample with a reagent which specifically interacts with the polynucleotide or the proteine kinase-like protein polypeptide.
9. A diagnostic kit for conducting the method of any one of claims 6 to 8.
10. A method of screening for agents which decrease the activity of a proteine kinase-like protein, comprising the steps of:
contacting a test compound with any proteine kinase-like protein polypeptide encoded by any polynucleotide of claim 1;
detecting binding of the test compound to the proteine kinase-like protein polypeptide, wherein a test compound which binds to the polypeptide is identified as a potential therapeutic agent for decreasing the activity of a proteine kinase-like protein.
11. A method of screening for agents which regulate the activity of a proteine kinase-like protein, comprising the steps of:
contacting a test compound with a proteine kinase-like protein polypeptide encoded by any polynucleotide of claim 1; and
detecting a proteine kinase-like protein activity of the polypeptide, wherein a test compound which increases the proteine kinase-like protein activity is identified as a potential therapeutic agent for increasing the activity of the proteine kinase-like protein, and wherein a test compound which decreases the proteine kinase-like protein activity of the polypeptide is identified as a potential therapeutic agent for decreasing the activity of the proteine kinase-like protein.
12. A method of screening for agents which decrease the activity of a proteine kinase-like protein, comprising the steps of:
contacting a test compound with any polynucleotide of claim 1 and detecting binding of the test compound to the polynucleotide, wherein a test compound which binds to the polynucleotide is identified as a potential therapeutic agent for decreasing the activity of proteine kinase-like protein.
13. A method of reducing the activity of proteine kinase-like protein, comprising the steps of:
contacting a cell with a reagent which specifically binds to any polynucleotide of claim 1 or any proteine kinase-like protein polypeptide of claim 4, whereby the activity of proteine kinase-like protein is reduced.
14. A reagent that modulates the activity of a proteine kinase-like protein polypeptide or a polynucleotide wherein said reagent is identified by the method of any of the claim 10 to 12.
15. A pharmaceutical composition, comprising:
the expression vector of claim 2 or the reagent of claim 14 and a pharmaceutically acceptable carrier.
16. Use of the expression vector of claim 2 or the reagent of claim 14 in the preparation of a medicament for modulating the activity of a proteine kinase-like protein in a disease.
17. Use of claim 16 wherein the disease is cancer, a CNS disorder, COPD, obesity, diabetes, or a cardiovascular disorder.
18. A cDNA encoding a polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9.
19. The cDNA of claim 18 which comprises SEQ ID NOS:1, 8 or 10.
20. The cDNA of claim 18 which consists of SEQ ID NOS:1, 8 or 10.
21. An expression vector comprising a polynucleotide which encodes a polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9.
22. The expression vector of claim 21 wherein the polynucleotide consists of SEQ ID NOS:1, 8 or 10.
23. A host cell comprising an expression vector which encodes a polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9.
24. The host cell of claim 23 wherein the polynucleotide consists of SEQ ID NOS:1, 8 or 10.
25. A purified polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9.
26. The purified polypeptide of claim 25 which consists of the amino acid sequence shown in SEQ ID NOS:2 or 9.
27. A fusion protein comprising a polypeptide having the amino acid sequence shown in SEQ ID NOS:2 or 9.
28. A method of producing a polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9, comprising the steps of:
culturing a host cell comprising an expression vector which encodes the polypeptide under conditions whereby the polypeptide is expressed; and
isolating the polypeptide.
29. The method of claim 28 wherein the expression vector comprises SEQ ID NOS:1, 8 or 10.
30. A method of detecting a coding sequence for a polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9, comprising the steps of:
hybridizing a polynucleotide comprising 11 contiguous nucleotides of SEQ ID NOS:1, 8 or 10 to nucleic acid material of a biological sample, thereby forming a hybridization complex; and
detecting the hybridization complex.
31. The method of claim 30 further comprising the step of amplifying the nucleic acid material before the step of hybridizing.
32. A kit for detecting a coding sequence for a polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9, comprising:
a polynucleotide comprising 11 contiguous nucleotides of SEQ ID NOS:1, 8 or 10; and
instructions for the method of claim 30.
33. A method of detecting a polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9, comprising the steps of:
contacting a biological sample with a reagent that specifically binds to the polypeptide to form a reagent-polypeptide complex; and
detecting the reagent-polypeptide complex.
34. The method of claim 33 wherein the reagent is an antibody.
35. A kit for detecting a polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9, comprising:
an antibody which specifically binds to the polypeptide; and
instructions for the method of claim 33.
36. A method of screening for agents which can modulate the activity of a human proteine kinase-like protein, comprising the steps of:
contacting a test compound with a polypeptide comprising an amino acid sequence selected from the group consisting of: (1) amino acid sequences which are at least about 35% identical to the amino acid sequence shown in SEQ ID NOS:2 or 9 and (2) the amino acid sequence shown in SEQ ID NOS:2 or 9; and
detecting binding of the test compound to the polypeptide, wherein a test compound which binds to the polypeptide is identified as a potential agent for regulating activity of the human proteine kinase-like protein.
37. The method of claim 36 wherein the step of contacting is in a cell.
38. The method of claim 36 wherein the cell is in vitro.
39. The method of claim 36 wherein the step of contacting is in a cell-free system.
40. The method of claim 36 wherein the polypeptide comprises a detectable label.
41. The method of claim 36 wherein the test compound comprises a detectable label.
42. The method of claim 36 wherein the test compound displaces a labeled ligand which is bound to the polypeptide.
43. The method of claim 36 wherein the polypeptide is bound to a solid support.
44. The method of claim 36 wherein the test compound is bound to a solid support.
45. A method of screening for agents which modulate an activity of a human proteine kinase-like protein, comprising the steps of:
contacting a test compound with a polypeptide comprising an amino acid sequence selected from the group consisting of: (1) amino acid sequences which are at least about 35% identical to the amino acid sequence shown in SEQ ID NOS:2 or 9 and (2) the amino acid sequence shown in SEQ ID NOS:2 or 9; and
detecting an activity of the polypeptide, wherein a test compound which increases the activity of the polypeptide is identified as a potential agent for increasing the activity of the human proteine kinase-like protein, and wherein a test compound which decreases the activity of the polypeptide is identified as a potential agent for decreasing the activity of the human proteine kinase-like protein.
46. The method of claim 45 wherein the step of contacting is in a cell.
47. The method of claim 45 wherein the cell is in vitro.
48. The method of claim 45 wherein the step of contacting is in a cell-free system.
49. A method of screening for agents which modulate an activity of a human proteine kinase-like protein, comprising the steps of:
contacting a test compound with a product encoded by a polynucleotide which comprises the nucleotide sequence shown in SEQ ID NOS:1, 8 or 10; and
detecting binding of the test compound to the product, wherein a test compound which binds to the product is identified as a potential agent for regulating the activity of the human proteine kinase-like protein.
50. The method of claim 49 wherein the product is a polypeptide.
51. The method of claim 49 wherein the product is RNA.
52. A method of reducing activity of a human proteine kinase-like protein, comprising the step of:
contacting a cell with a reagent which specifically binds to a product encoded by a polynucleotide comprising the nucleotide sequence shown in SEQ ID NOS:1, 8 or 10, whereby the activity of a human proteine kinase-like protein is reduced.
53. The method of claim 52 wherein the product is a polypeptide.
54. The method of claim 53 wherein the reagent is an antibody.
55. The method of claim 52 wherein the product is RNA.
56. The method of claim 55 wherein the reagent is an antisense oligonucleotide.
57. The method of claim 56 wherein the reagent is a ribozyme.
58. The method of claim 52 wherein the cell is in vitro.
59. The method of claim 52 wherein the cell is in vivo.
60. A pharmaceutical composition, comprising:
a reagent which specifically binds to a polypeptide comprising the amino acid sequence shown in SEQ ID NOS 2 or 9; and
a pharmaceutically acceptable carrier.
61. The pharmaceutical composition of claim 60 wherein the reagent is an antibody.
62. A pharmaceutical composition, comprising:
a reagent which specifically binds to a product of a polynucleotide comprising the nucleotide sequence shown in SEQ ID NOS:1, 8 or 10; and
a pharmaceutically acceptable carrier.
63. The pharmaceutical composition of claim 62 wherein the reagent is a ribozyme.
64. The pharmaceutical composition of claim 62 wherein the reagent is an antisense oligonucleotide.
65. The pharmaceutical composition of claim 62 wherein the reagent is an antibody.
66. A pharmaceutical composition, comprising:
an expression vector encoding a polypeptide comprising the amino acid sequence shown in SEQ ID NOS:2 or 9; and
a pharmaceutically acceptable carrier.
67. The pharmaceutical composition of claim 66 wherein the expression vector comprises SEQ ID NOS:1, 8 or 10.
68. A method of treating a proteine kinase-like protein dysfunction related disease, wherein the disease is selected from cancer, a CNS disorder, COPD, obesity, diabetes, or a cardiovascular disorder comprising the step of:
administering to a patient in need thereof a therapeutically effective dose of a reagent that modulates a function of a human proteine kinase-like protein, whereby symptoms of the proteine kinase-like protein disfunction related disease are ameliorated.
69. The method of claim 68 wherein the reagent is identified by the method of claim 36.
70. The method of claim 68 wherein the reagent is identified by the method of claim 45.
71. The method of claim 68 wherein the reagent is identified by the method of claim 49.
US10/471,762 2001-03-16 2002-03-15 Regulation of human protein kinase-like protein Expired - Fee Related US7148050B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/471,762 US7148050B2 (en) 2001-03-16 2002-03-15 Regulation of human protein kinase-like protein

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US27605501P 2001-03-16 2001-03-16
US32405301P 2001-09-24 2001-09-24
US32645801P 2001-10-03 2001-10-03
US33712401P 2001-12-10 2001-12-10
US10/471,762 US7148050B2 (en) 2001-03-16 2002-03-15 Regulation of human protein kinase-like protein
PCT/EP2002/002887 WO2002081704A2 (en) 2001-03-16 2002-03-15 Regulation of human protein kinase-like protein

Publications (2)

Publication Number Publication Date
US20040171539A1 true US20040171539A1 (en) 2004-09-02
US7148050B2 US7148050B2 (en) 2006-12-12

Family

ID=29587834

Family Applications (1)

Application Number Title Priority Date Filing Date
US10/471,762 Expired - Fee Related US7148050B2 (en) 2001-03-16 2002-03-15 Regulation of human protein kinase-like protein

Country Status (7)

Country Link
US (1) US7148050B2 (en)
EP (1) EP1373487B1 (en)
AT (1) ATE363529T1 (en)
AU (1) AU2002315253A1 (en)
DE (1) DE60220388T2 (en)
ES (1) ES2287285T3 (en)
WO (1) WO2002081704A2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5635177A (en) * 1992-01-22 1997-06-03 Genentech, Inc. Protein tyrosine kinase agonist antibodies
US20040053394A1 (en) * 2001-10-20 2004-03-18 Rajagopal Gururajan Human kinases

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003512044A (en) 1999-10-15 2003-04-02 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド Protein tyrosine kinase receptor polynucleotides, polypeptides, and antibodies
EP1261636A2 (en) * 2000-03-06 2002-12-04 Sugen, Inc. Human protein kinases and protein kinase-like enzymes
CA2425963A1 (en) 2000-10-20 2002-04-25 Incyte Genomics, Inc. Human kinases
EP1260519A1 (en) * 2001-04-26 2002-11-27 Schering Aktiengesellschaft Membrane receptors for steroids and sterols

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5635177A (en) * 1992-01-22 1997-06-03 Genentech, Inc. Protein tyrosine kinase agonist antibodies
US20040053394A1 (en) * 2001-10-20 2004-03-18 Rajagopal Gururajan Human kinases

Also Published As

Publication number Publication date
DE60220388D1 (en) 2007-07-12
WO2002081704A2 (en) 2002-10-17
ATE363529T1 (en) 2007-06-15
US7148050B2 (en) 2006-12-12
EP1373487B1 (en) 2007-05-30
DE60220388T2 (en) 2008-01-24
AU2002315253A1 (en) 2002-10-21
ES2287285T3 (en) 2007-12-16
WO2002081704A3 (en) 2003-04-10
EP1373487A2 (en) 2004-01-02

Similar Documents

Publication Publication Date Title
US20040038365A1 (en) Regulation of human lysosomal acid lipase
US20040241156A1 (en) Regulation of human aminopeptidase n
US7049118B2 (en) Regulation of human serine-threonine protein kinase
EP1360281B1 (en) Regulation of human wee1-like serine/threonine protein kinase
US20040241796A1 (en) Regulation of human nek-like serine/threonine protein kinase
US20040152092A1 (en) Regulation of human phosphatidic acid phosphatase type 2c-like protein
US7148050B2 (en) Regulation of human protein kinase-like protein
US7150976B2 (en) Regulation of human serine-threonine protein kinase
US20040136976A1 (en) Regulation of human zinc carboxypeptidase b-like protein
WO2002055710A2 (en) Regulation of human purple acid phosphatase
US20040235094A1 (en) Regulation of human ptpase-like protein
WO2003018786A2 (en) Human serine/threonine kinase
WO2003033709A2 (en) Regulation of human serine/threonine protein kinase
US20040137593A1 (en) Regulation of human serine/threonine protein kinase-like protein
US20040157282A1 (en) Regulation of human dual specificity protein phosphatase 7-like protein
WO2003000873A2 (en) Regulation of human serine/threonine protein kinase nek1
WO2003000874A2 (en) Regulation of human serine/threonine protein kinase nek3
WO2002083887A2 (en) Regulation of human methionine aminopeptidase-like protein
WO2002090543A2 (en) Regulation of human phosphatidic acid phosphatase type 2c-like protein
WO2002038776A1 (en) Regulation of human fatty acid coa ligase
WO2002022791A2 (en) Regulation of human pyridoxine 5&#39;-phosphate oxidase
WO2003035859A2 (en) Regulation of human serine/threonine kinase
WO2002070678A2 (en) Regulation of human serine/threonine protein kinase
WO2002099094A2 (en) Human serine/threonine protein kinase-like enzyme
WO2002083904A1 (en) Regulation of human protein phosphatase 2c-like protein

Legal Events

Date Code Title Description
AS Assignment

Owner name: BAYER AKTIENGESELLSCHAFT, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SMOLYAR, ALEX;REEL/FRAME:015254/0562

Effective date: 20030909

AS Assignment

Owner name: BAYER HEALTHCARE AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BAYER AKTIENGESELLSCHAFT;REEL/FRAME:018431/0302

Effective date: 20061023

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: BAYER SCHERING PHARMA AG, GERMANY

Free format text: MERGER;ASSIGNOR:BAYER HEALTHCARE AG;REEL/FRAME:022277/0324

Effective date: 20081204

FPAY Fee payment

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Free format text: PAYER NUMBER DE-ASSIGNED (ORIGINAL EVENT CODE: RMPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20141212